Next Article in Journal
Type 2 Diabetes Related Mitochondrial Defects in Peripheral Mononucleated Blood Cells from Overweight Postmenopausal Women
Next Article in Special Issue
Extracorporeal Carbon Dioxide Removal: From Pathophysiology to Clinical Applications; Focus on Combined Continuous Renal Replacement Therapy
Previous Article in Journal
Implementing Translational Research to Understand the Future of COVID-19 and Its Long-Term Consequences: A Degrowth Perspective or the Transformation of a Global Emergency?
Previous Article in Special Issue
Development and Characterization of Phage-Display-Derived Novel Human Monoclonal Antibodies against the Receptor Binding Domain of SARS-CoV-2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Stem Cell Therapy for Alzheimer’s Disease: A Scoping Review for 2017–2022

1
Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
2
Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT 06510, USA
3
Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
4
Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, China
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(1), 120; https://doi.org/10.3390/biomedicines11010120
Submission received: 28 October 2022 / Revised: 28 December 2022 / Accepted: 28 December 2022 / Published: 3 January 2023

Abstract

:
Alzheimer’s disease (AD) has been a major causal factor for mortality among elders around the world. The treatments for AD, however, are still in the stage of development. Stem cell therapy, compared to drug therapies and many other therapeutic options, has many advantages and is very promising in the future. There are four major types of stem cells used in AD therapy: neural stem cells, mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells. All of them have applications in the treatments, either at the (1) cellular level, in an (2) animal model, or at the (3) clinical level. In general, many more types of stem cells were studied on the cellular level and animal model, than the clinical level. We suggest for future studies to increase research on various types of stem cells and include cross-disciplinary research with other diseases. In the future, there could also be improvements in the timeliness of research and individualization for stem cell therapies for AD.

1. Introduction

1.1. Alzheimer’s Disease and Its Treatments

Among elderly people around the world, Alzheimer’s disease (AD) is one of the major causal factors for mortality [1]. AD is a progressive chronic disease that causes a degenerative disorder of the brain, which gradually worsens memory and eventually leads to disability in performing communication and basic daily tasks [2].
Up until now, numerous approaches to therapeutic drugs are encountering unsatisfying outcomes in improving the cognitive performance of Alzheimer’s disease [3]. The reason why drugs are not curing it well is very complex. One of the reasons is unclear pathology. In other words, the pathogenesis of AD is still under investigation [4]. Another reason is that AD is a chronic disease that requires long-term care. This is unlike treatment for acute diseases where short-term drug therapy is viable, and the outcome is timely. As time progresses, the method of drug intake for AD patients needs careful modification [5]. Most importantly, therapeutic drugs are not able to stimulate the regeneration of neural cells that are already damaged. As the level of severity of AD increases, the lack of vitality of body cells also inhibits the effective transportation of drug molecules [6]. Thus, the efficiency and accuracy of drug therapy are highly restrained.
Besides drugs, most other therapeutic options have not yet shown significant effectiveness in treating AD. Brain-derived neurotrophic factors, despite their importance in maintaining synaptic plasticity in memory and learning, have not yet been discovered in terms of their exact signaling mechanisms [7]. Insulin therapy, despite its potential in safe and short-term symptomatic intervention to delay cognition loss, does not always show significant clinical outcomes in real-life settings [8]. Low-level laser therapy can potentially prevent cognitive impairment by altering brain cell function and neurometabolic pathways, but the optimal wavelength, dosage, and intensity have not been determined for individual treatment. The duration of effects and the chance of chronic repetition are also unclear [9]. Herbal medicine has shown mild effectiveness in treating cognitive deficits of AD, but the action mechanisms of herbs and most of their biochemical formulations are not clearly understood. In addition, since herbal medicines are from natural products, the content and concentration of chemical constituents vary from batch to batch, and could be difficult to standardize [10]. Mitochondrial calcium targeting has the potential to achieve more than symptomatic relief because mitochondrial respiration plays a crucial role in AD etiology [11]. However, why mitochondrial calcium efflux is downregulated early in AD pathogenesis is still a question [12]. Signaling pathways have the potential to provide multilevel, multifaceted, and multitargeted approaches to prevent and treat AD, but it is challenging to implement them in clinical settings due to their complexity [13]. A healthy lifestyle, including moderate physical activity and an appropriate diet, also has an association with lowering the risk of AD. Yet, it is not as effective in the treatment of AD [14].
However, stem cell therapy, which has been developed relatively recently, gives hope for better treatment of AD. Stem cell therapy enhances the level of functional recovery in the central nervous system of the brain [15]. By implementing exogenous stem cells, the depleted neuronal circuitry could be repopulated and regenerated [16]. Stem cell therapy is able to reduce neuroinflammation, which is especially important for patients who develop AD after aneurysmal subarachnoid hemorrhage (SAH) because neuroinflammation plays a vital role in injury expansion and brain damage that eventually cause cognitive decline [17]. Stem cell therapy can also eliminate neurofibrillary tangles and abnormal degradation of proteins, and promote mitochondrial transport to improve cognition [18]. In particular, in the early stages of AD, neural stem cells are able to participate extensively in brain homeostasis, which repairs and exhibits pleiotropic intrinsic properties to mitigate and eventually cure AD [19]. Stem cell-derived exosomes can also have donor-derived properties and minimal immunogenicity, which lower the risk of tumor formation after therapy [20].

1.2. Types of Stem Cell for AD Therapy

To thoroughly understand the mechanisms of stem cell therapy for AD, we will evaluate therapies from various types of stem cells and review their development respectively. In general, there are four major types of stem cells used for AD therapy: (1) neural stem cells (2) mesenchymal stem cells (3) embryonic stem cells (4) induced pluripotent stem cells [21].

1.2.1. Neural Stem Cell (NSC)

Up until now, the regeneration of cognitive decline and loss of brain tissue in AD patients have been non-curable. Thus, the majority of effective AD therapies focus on targeting AD pathology in the early stage to preserve cerebrovascular function. Because NSCs contribute considerably to brain homeostasis and repair, they reveal pleiotropic fundamental properties for the treatment of AD in the early stages [19].
In order to develop treatments for AD, it is vital to develop experimental models which represent a specific cellular phase of AD and laborious analysis of the cellular pathological mechanisms [22]. In 2018, McGinley et al. [23] discovered that transplantation of human NSC enhanced cognition of AD in a murine model APP/PS1 (amyloid precursor protein and presenilin 1 mutated mice). The transplantation was targeted to the fimbria fornix, and it significantly improved cognition in the hippocampal-dependent memory tasks at 4 and 16 weeks after transplantation. Furthermore, in 2020, Hayashi et al. [24] modeled human-derived NCS (hNSC) and murine-derived NSC (mNSC) transplantation. Both hNSC and mNSC gave positive results in treating AD.
More recent research has dived into the cellular mechanisms of NSC, and its therapeutic pathology for AD. In 2021, Apodaca et al. [25] discovered that hNSC-derived extracellular vesicles can mitigate the hallmarks of AD. They gave 2-/6-month old 5 × AD mice injections of hNSC-derived extracellular vesicles (EV). NSC treatment significantly decreased dense core amyloid-β plaque accumulation in both age groups, which showed neuroprotective effects for the redress of AD neuropathologies. In 2022, Reveulta et al. [26] studied microglia-mediated inflammation and NSC differentiation in AD, and the possible therapeutic effect of K(V)1.3 channel blockade. They concluded that K(V)1.3 blockers hinder microglia-mediated neurotoxicity in culture, reducing the manifestation and construction of the pro-inflammatory cytokines through NF-kB and p38MAPK pathways.
In general, NSC therapy has developed more advanced and detailed pathology mechanisms, with greater effectiveness in treating AD in the early stage.

1.2.2. Mesenchymal Stem Cell (MSC)

MSC is the most studied type of stem cell in stem cell therapies for AD, due to its excellent accessibility and wide range of differentiating potential. It can be administered intravenously to perform blood–brain barrier penetration with low immune response. In particular, MSC-derived exosomes (MSC-exos) are able to have donor-derived properties with minimal immunogenicity. MSC-exos also have little risk of forming tumors post-therapy, which make them a promising treatment for AD [20].
Several pre-clinical research studies have received significant results in recent years. In 2019, Zaldivar et al. [27] discovered that MSC-exos could increase neural plasticity and enhance cognitive impairment. They injected amyloid-β 1–42 aggregates into the dentate gyrus of murine models bilaterally, and performed novel object recognition tests on days 14 and 28. Results indicated that MSC-exos stimulated neurogenesis in the subventricular zone. In 2020, Nakano et al. [28] discovered that bone marrow-derived MSC (BM-MSC) could enhance cognitive impairment in an AD model by enhancing the expression of microRNA-146a in the hippocampus. BM-MSC were injected intracerebroventricularly into the choroid plexus in the lateral ventricle, and secreted exosomes in the cerebrospinal fluid. In vitro experiments illustrated that exosomal miR-146a from BM-MSC was absorbed in astrocytes, and the level of miR-146a was increased. As the key to forming synapses, astrocytes restore cognitive function and mitigate AD. In the same year, Wei et al. [29] also investigated whether MSC-derived exosomal miR-223 regulates apoptosis of neuronal cells. MSC-derived exosomal miR-223 targeted PTEN, thus activating the PI3K/Akt pathway to inhibit neuron apoptosis, and hence become a potential treatment for AD.
Despite extensive pre-clinical research, research on the clinical level has also made remarkable progress recently. In 2021, Kim et al. [30] performed an intracerebroventricular injection of human umbilical cord blood MSC (hUCB-MSC) in AD patients in a phase I clinical trial. They recruited nine mild-to-moderate AD patients and injected low and high doses of hUCB-MSC, respectively. All adverse events subsided within 36 h, and their symptoms of AD were mitigated.
Overall, MSC therapy reduces neuroinflammation by eliminating amyloid-β, tangles in neuro fibers, and abnormal degradation of proteins. MSC therapy promotes blood–brain barrier and autophagy-related recoveries, regulates acetylcholine levels, and improves cognition of the brain [18].

1.2.3. Embryonic Stem Cell (ESC)

Because there are still ethical and immunogenic limitations to using ESC for treating AD [18], clinical implementation of ESC-based therapy may not be applicable in the short-term. However, there have been a few pre-clinical studies that have shown progress in using ESC to model AD pathology.
On the genetic and cellular level, ESC modeling has made much progress. In 2019, Ubina et al. [31] modeled human ESC on Aβ-dependent neurodegeneration. An allele of APP locus was modified to express Aβ40/Aβ42 secretory so that the edited allele expression could pass the amyloidogenic APP processing pathway. After neural differentiation, pathway analysis indicated downregulation of the extracellular matrix and over-expression in cilia functions. In 2021, Fan et al. [32] discovered that SIRT1 controls sphingolipid metabolism and neural differentiation of ESC through c-Myc-SMPDL3B. They focused on sphingolipids because they are vital structures of the cell membrane, which regulate cell differentiation and apoptosis. In AD patients, there is a deficit in creating enzymes to remove excess levels of sphingolipids, which eventually leads to neurodegeneration. In particular, SIRT1, an NAD(+)-dependent protein deacetylase, regulates the degradation of sphingolipids by increasing the production of the enzyme SMPDL3B. Therefore, targeting SIRT1 may offer innovative strategies to treat AD. Notably, SIRT1 is sensitive to high-fat diets; therefore, maternal obesity could be a cause of AD as infants develop into adulthood.
In addition, there are also studies with animal models on ESCs. In 2020, Kim et al. [33] investigated the efficacy and feasibility of intra-arterial administration of ESC in an animal model of AD. MSC significantly inhibited Aβ-induced cell apoptosis in the hippocampus, and increased autophagolysosomal clearance of Aβ. MSC-treated mice performed with higher memory ability than those with only Aβ injection.

1.2.4. Induced Pluripotent Stem Cell (iPSC)

iPSC is a technology in which somatic cells are reprogrammed to pluripotent stem cells, creating an optimal physiologically relevant model that maintains the donor’s genetic identity. iPSC can unlimitedly self-renew in vitro, and differentiate into various cell types, which gives hope to model and cure AD [34].
On the genetic and cellular level, there have been multiple studies on iPSC therapy for AD. In 2020, Butler et al. [35] discovered the genetic relevance of human iPSC-derived microglia (iMG) to AD. Microglia are the major immune cell in the brain that imply the pathogenesis of AD. Using gene expression specific to cell type, they showed that iMG cells are genetically relevant to AD. In 2020, Zhang et al. [36] found that human iPSC-derived neural cells from AD patients showed various susceptibilities to oxidative stress. The oxidative stress response of neural cells is a vital mechanism for cognitive dysfunction and aging in AD. Under exposure to H2O2, the vitality and neurite length of human iPSC-induced neurons reduced significantly. Due to the oxidative property of neuron cells, there is a potential to treat AD by targeting the de-oxidization of the neurons.
In a mouse model of AD, iPSC-derived neural precursors showed improvement in memory and synaptic abnormalities. Researchers injected mouse iPSC-derived neural precursors (iPSC-NPCs) stereotaxically into the hippocampus of mice. Mice with iPSC-NPCs transplantation revealed improvement in synaptic plasticity and reduced AD brain pathology, including decreased tangles deposits and amyloid [37].
iPSC can also be used for drug screening and testing for AD. The flexibility of iPSC includes non-invasive harvesting compatibility and sourcing from patients with AD [38]. In addition, compared to ESC as we discussed in the Section 1.2.3, iPSC is able to reprogram cells without embryo destruction and with negligibly invasive processes [39]. iPSC could create neuronal cells from specific patients, and eradicate the drawback of species-specificity inherent when using animal models. There are also a few novel technologies that can be combined with iPSC models to treat AD, including organoid technology, genome editing, deep learning artificial intelligence, and single-cell RNA sequencing [40].

1.3. Overview of Study and Objective

Despite the promising future of stem cell therapy on AD, most research remains in the pre-clinical stage [41]. Therefore, it is worthwhile investigating why there is a gap between animal models and clinical applications, and how to improve it.
This scoping review was conducted to map the research completed in this area systematically, and to identify any existing gaps in knowledge. The following research question was formulated:
‘How did the different types of stem cells used for Alzheimer’s disease in studies on the cellular level, animal model, and clinical level imply their effectiveness?’

2. Methodology

2.1. Search Strategy

This scoping review follows the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [42]. The review was performed as described in Figure 1, including papers extracted from PubMed, Web of Science, EMBASE, and Scopus. The papers were intentionally selected to be published in the range of years from 2017 to 2022, i.e., the most recent 5 years, in order to evaluate stem cell therapy’s effectiveness on Alzheimer’s disease using up-to-date data.
The following search query terms were used in the searching databases:
((((Alzheimer[Title/Abstract]) OR (AD[Title/Abstract]) OR (MCI[Title/Abstract]) OR (mild cognitive impairment[Title/Abstract])) AND (stem cell[Title/Abstract]) AND ((therapy[Title/Abstract]) OR (treatment[Title/Abstract]) OR (medication[Title/Abstract]))) NOT (Review)) NOT (Parkinson).
The search was completed in October 2022.
During data selection and review, the search of literature was completed by two researchers in independent locations, and they completed the prepared results. When there was any disagreement between the researchers during the process, they would review the main text together in a research meeting to achieve a consensus result.

2.2. Inclusion and Exclusion Criteria

Studies on stem cell therapy for Alzheimer’s disease from 2017 to 2022 were included. Both stem cell therapy and AD had to be mentioned in the article for the studies to be eligible. To be included, the studies also needed to address the therapeutic effects of stem cells and how they relate to the treatment of AD.
After screening and removing duplicated articles, 179 full-text articles were assessed for eligibility, and 117 of them were excluded for the following three reasons:
  • Reason 1 (n = 41): “stem cell” was mentioned in the article, not as a means of therapy but as a medium for other medical treatments or preclinical research. For instance, Kim et al. [43] mentioned induced pluripotent stem cells (iPSCs) only as a model for AD to aid their research on estrogen’s mitigating effect on AD. Even though this kind of research was related to stem cells, it did not have a significant relationship with stem cell therapy and thus was excluded from our records.
  • Reason 2 (n = 42): the article contained brief mentioning of AD, but was not targeted toward AD. For instance, Rajan et al. [44] mentioned a collection of neurodegenerative diseases, with AD just as a sub-section of the entire article. Even though this article explained well the mechanisms of stem cell therapy, it did not include enough information for the treatment of AD and thus could not be included in our records.
  • Reason 3 (n = 34): the exposure group (i.e., treated with stem cell therapy) could not be qualified as eligible to be included in the synthesis. For instance, Campos et al. [45] compared two types of stem cell therapies (neural and mesenchymal), instead of one with a control group. Therefore, it is difficult to qualify and quantify the effects of their research.
Overall, 62 studies were included in the qualitative synthesis. Of the 62 articles, 12 were research on the cellular level, 43 were research on an animal model, and 7 were research on the clinical level.

3. Analysis of the Included Studies

In Figure 2, we summarized the distribution (number of studies) for different types of stem cells on (Figure 2a) cellular level, (Figure 2b) animal model, (Figure 2c) clinical level. Clearly, there were many more types of stem cells included in animal model studies than at the cellular level and clinical level. The aggregate number of studies was also greater in animal model studies.
In Figure 3, we collected the effects of different types of stem cells on treating AD, including studies from cellular level, animal model, and clinical level. The goal of all was to mitigate and ultimately cure AD. To achieve this, the research, with different types of stem cells, showed variety in levels of progress. In particular, MSCs showed the most significant effects due to their popularity. The types of stem cells are listed according to their effectiveness from the top (MSC being the most effective) to the bottom (several other types of stem cells).
In the following sections, we are going to present the details of studies on cellular level, animal model, and clinical level, respectively.

3.1. Cellular Level

In general, studies on the cellular level of stem cell therapy for AD centered on three types of stem cells: MSC, iPSC, and NSC. Among them, iPSC is the most studied type of stem cell. Most notably, modeling with iPSCs derived from AD patients showed the significance of Apolipoprotein E4 (APOE4) variant as a risk factor for AD. [46,47] MSC is the second most studied type of stem cell among cellular level studies, and there are various sub-types of MSCs in the studies, including: BM-MSC (bone-marrow mesenchymal stem cell), ucMSC (human umbilical cord mesenchymal stem cell), hUCB-MSC (human umbilical cord blood derived mesenchymal stem cell), and hucMSC (human umbilical cord derived mesenchymal stem cell). ucMSC, hUCB-MSC, and hucMSC refer to the same type of stem cell, but we named them differently to show respect for the nomenclature used in different studies. Every stem cell type starts with an “h” referring to a human-oriented stem cell, for instance hNSC is human NSC, and hiPSC is human iPSC. The types of stem cells were also categorized according to their orientation. Among the 12 studies, 1 is a study with animal-induced stem cells, and 11 are studies with human-oriented stem cells. Table 1 is a detailed summary of the control group, intervention, and measured outcomes of interest for studies on the cellular level.

3.2. Animal Model

There are many more studies and types of stem cells included in our record of studies for a murine model than on the cellular level. A murine model is the most widely used animal model for studies on stem cell therapy for AD. The benefit of using an animal model is the ability to evaluate the effectiveness of therapies on a tissue level, and this case the brain. A murine model also allows for the conduction of experiments with a larger and more flexible sample size than in clinical research, with minimal concern of ethical issues.
Among various types of stem cells, MSC is the most prevalent type, including many subtypes. sRAGE-MSC is a soluble receptor for an advanced glycation end-products derived mesenchymal stem cell; ES-MSC is ESC derived MSC; AD-MSC is adipose-derived MSC. MFSCE is a membrane-free stem cell, which is a component of adipose-tissue-derived stem cells, where ATSC is an adipose tissue mesenchymal stem cell and hADSC is a human adipose-derived stem cell. In addition to MSCs, there are a variety of other types of stem cells. SCF refers to stem cell factor; hNTSC is human neural crest-derived nasal turbinate stem cell, and SHED is a stem cell from human exfoliated deciduous teeth.
Table 2 is a detailed summary of the control group, intervention, and measured outcomes of interest for the animal model studies.

3.3. Clinical Level

In our records, the published results of stem cell therapy for AD on the clinical level, from 2017 to 2022, were much less than the number of those for the animal model. Most clinical-level studies include iPSC because stem cells were generated from specific patients. Table 3 is a detailed summary of the control group, intervention, and measured outcomes of interest for studies on the clinical level.

4. Discussion

In our review, we summarized various types of stem cell therapies for AD in the time period from 2017 to 2022. We categorized studies according to whether they were on the cellular level, animal model, or clinical level. We did so to visualize the abundance of research at the pre-clinical level, compared to the deficiency in mature clinical level experiments. The reason behind massive pre-clinical studies is the unclear pathology for AD with stem cell therapy, despite its potential effectiveness. If the pathology is clear and acknowledged, there should be more clinical-level experiments than pre-clinical studies because then studies would move forward to applications in clinical settings. Therefore, the current primary focus of research should be on exploring effective pathologies for potential treatments, and clinical level studies would provide support for the pre-clinical studies.
Among pre-clinical studies, we discovered that MSCs had the maximum quantity of studies, and the most significant effect. Two remarkable effects of MSCs are (1) increase in telomerase activity, and (2) decrease in tau phosphorylation, which restored hippocampal neuronal morphology and improved brain glucose metabolism. MSC is also the only type of stem cell used in clinical level studies that progressed to phase I clinical trials. While iPSC also has applications on the clinical level, it is a reversed approach that generates stem cells from AD patients to aid in analysis based on patient-generated stem cell genetics. Other types of stem cells showed potential for treatments in pre-clinical studies, but not in clinical level studies. Therefore, we suggest future researchers to explore clinical outcomes of MSCs, and expand on the types of other stem cells in the pre-clinical stage. These types of stem cells could include neural crest stem cells, hematopoietic stem cells, and so on [41].
We also encourage cross-disciplinary research with other diseases. Stem cell therapy can not only treat AD, but also many other types of disease such as cancer. In particular, one study has shown that cancer and AD are at opposite sides of the cell division spectrum, and hence cannot happen at the same time [102]. Using this feature, scientists can potentially lower the side effects of stem cell therapy on AD patients who have the risk of developing tumors.
In addition, researchers could create a loop of response from pre-clinical studies and the medical record system in a timely manner, in order to stimulate the efficiency of feedback for therapeutic methods. In other words, there should be instant communication between pre-clinical research and clinical data, without active searching from researchers or physicians. When we used the search engines such as PubMed, they displayed all research in the field according to relevance and time, but did not show a map of correlation between individual experiments. Additionally, since AD is a chronic disease, the time span is relatively long compared to other acute diseases. It would be crucial to track the timing of treatments to find the optimal proportionality of therapeutic injection for each stage of AD. When there are outliers in research or clinical data that are not in-line with therapeutic expectations, secondary-level analysis can be performed on the frequency of outlier occurrence chronologically to track the potential causes, and to contribute to better cure methods and prevention.
Despite the potential effectiveness of stem cell therapy for AD, there are also a few limitations regarding each specific type of stem cell. First, there is a lack of application of NSCs to large-scale clinical trials in AD [103] because isolation and enough expansion of NSCs from the central nervous system are difficult in vitro, and require culture medium supplemented with mitogenic growth factors, such as epidermal growth factor and basic fibroblast growth factor [104]. For iPSCs, there is risk of tumorigenicity and infection, especially in derived donor cells associated with iPSCs-based therapy for AD. These potential side effects bring about a lot of concerns from patients and physicians, especially when the cure for AD is not ascertained [105]. For ESCs, there are also side effects of tumor formation and graft failure, which could negatively impact the well-being of patients post therapy [106]. For MSCs, there is potential organelle dysfunction after therapy, which could be detrimental to the health of the patient [20]. In addition, there are also ethical issues and technical limitations in stem cell therapy. Current ethical concerns for stem cell therapy are centered on the unlimited differentiation potential of iPSCs, which can be used in human cloning [107]. Several technical limitations of stem cell therapy include immunogenicity and limited cell survival in vivo [108].
Individualization is also a key factor that prevents pre-clinical studies from being applied in clinical settings. For cell and animal models, there is less of a concern for the variation of each individual because the animal brain is much less complex than the human brain. For each human individual, however, the brain is much more sophisticated. Physicians also cannot track the memories of their patients because (1) the patients cannot recall everything regarding their previous life experiences, and (2) there is not a medical record of all the brain statuses or activities that can be referred to. A simple CT scan is sufficient for acute brain injuries, but not enough for long-term dementia-like AD. Therefore, the potential solution for this problem is first to ask each patient to evaluate their health situation and involve them in their treatment procedures so that physicians can get first-hand medical data, and second, to make their medical treatment (in this case stem cell therapy) flexible and adjustable as the development of AD grows more or less severe. Physicians and researchers should also consider the patient’s age, sex, family history, social background, and previous health records to make the treatment more suitable for their needs.
Furthermore, there are always concerns about the commercialization of stem cell therapy for AD because it would negatively influence the profits of already-existing drugs. However, these two therapies need not have absolute conflict. In one study, it is shown that iPSC and herbal small-molecule drugs can combine and treat AD more effectively [109]. Furthermore, stem cell therapy is currently not affordable for many patients with lower income, and it would be helpful to include it in medical insurance when the therapies are mature, so that the general public can afford it, and physicians can have more patients in their medical records to justify and improve pre-clinical research.
Above all, the strength of our review is the inclusion and combination of recent studies in this area systematically. Most previous reviews on stem cell therapies for AD had focused solely on: (1) pathology [110] (pre-clinical only) or (2) a specific type of stem cell (e.g., MSC only [111]); previous comprehensive reviews also comprised many articles that were not recent (i.e., within 5 years) [112]. Our scoping review includes recent studies on a variety of stem cells, and categorizes their pathological impacts on treating AD. However, the weakness of our review is the inability to include statistical analysis and quantitative comparisons. This is because the statistical methods in the included studies vary extensively, and it is difficult to standardize the results with specific criteria. Hopefully with more clinical studies emerging in the future there could be the potential to compare the results of studies by the standard of patient survival rate.

5. Conclusions

In conclusion, stem cell therapy is a promising treatment for Alzheimer’s disease (AD), but it is still in the process of development. There are four major types of stem cells for AD therapy: neural stem cells, mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells. All of them have applications in the studies of (1) cellular level, (2) animal model, and (3) clinical level of AD. In general, there were many more types of stem cells studied on the cellular level and in an animal model, than on the clinical level. We suggest future studies to increase research on various types of stem cells, and include cross-disciplinary research with other diseases. In the future, there could also be improvements in the timeliness of research and individualization for stem cell therapies for AD.

Author Contributions

Conceptualization, Y.D.; methodology, S.Z. and Y.D.; software, Y.D.; validation, Y.D.; formal analysis, Y.D. and L.L.; investigation, Y.D. and L.L.; resources, Y.D.; data curation, Y.D. and L.L.; writing—original draft preparation, Y.D.; writing—review and editing, Y.D. and S.Z.; visualization, Y.D.; supervision, S.Z.; project administration, Y.D. and S.Z.; funding acquisition, Y.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing does not apply to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Alipour, M.; Nabavi, S.M.; Arab, L.; Vosough, M.; Pakdaman, H.; Ehsani, E.; Shahpasand, K. Stem cell therapy in Alzheimer’s disease: Possible benefits and limiting drawbacks. Mol. Biol. Rep. 2019, 46, 1425–1446. [Google Scholar] [CrossRef]
  2. Bagheri-Mohammadi, S. Stem cell-based therapy as a promising approach in Alzheimer’s disease: Current perspectives on novel treatment. Cell Tissue Bank. 2021, 22, 339–353. [Google Scholar] [CrossRef] [PubMed]
  3. Bali, P.; Lahiri, D.K.; Banik, A.; Nehru, B.; Anand, A. Potential for Stem Cells Therapy in Alzheimer’s Disease: Do Neurotrophic Factors Play Critical Role? Curr. Alzheimer Res. 2017, 14, 208–220. [Google Scholar] [CrossRef] [PubMed]
  4. Agatonovic-Kustrin, S.; Kettle, C.; Morton, D.W. A molecular approach in drug development for Alzheimer’s disease. Biomed. Pharmacother. 2018, 106, 553–565. [Google Scholar] [CrossRef] [PubMed]
  5. Cummings, J.; Lee, G.; Ritter, A.; Sabbagh, M.; Zhong, K. Alzheimer’s disease drug development pipeline: 2020. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2020, 6, e12050. [Google Scholar] [CrossRef]
  6. Kabir, M.T.; Uddin, M.S.; Mamun, A.A.; Jeandet, P.; Aleya, L.; Mansouri, R.A.; Ashraf, G.M.; Mathew, B.; Bin-Jumah, M.N.; Abdel-Daim, M.M. Combination drug therapy for the management of Alzheimer’s disease. Int. J. Mol. Sci. 2020, 21, 3272. [Google Scholar] [CrossRef]
  7. Gao, L.; Zhang, Y.; Sterling, K.; Song, W. Brain-derived neurotrophic factor in Alzheimer’s disease and its pharmaceutical potential. Transl. Neurodegener. 2022, 11, 4. [Google Scholar] [CrossRef]
  8. Craft, S.; Raman, R.; Chow, T.W.; Rafii, M.S.; Sun, C.K.; Rissman, R.A.; Donohue, M.C.; Brewer, J.B.; Jenkins, C.; Harless, K.; et al. Safety, Efficacy, and Feasibility of Intranasal Insulin for the Treatment of Mild Cognitive Impairment and Alzheimer Disease Dementia: A Randomized Clinical Trial. JAMA Neurol. 2020, 77, 1099–1109. [Google Scholar] [CrossRef]
  9. De la Torre, J.C. Treating cognitive impairment with transcranial low level laser therapy. J. Photochem. Photobiol. B 2017, 168, 149–155. [Google Scholar] [CrossRef]
  10. Dos Santos-Neto, L.L.; de Vilhena Toledo, M.A.; Medeiros-Souza, P.; de Souza, G.A. The use of herbal medicine in Alzheimer’s disease-a systematic review. Evid.-Based Complement. Altern. Med. 2006, 3, 441–445. [Google Scholar] [CrossRef]
  11. Wu, A.J.; Tong, B.C.; Huang, A.S.; Li, M.; Cheung, K.H. Mitochondrial Calcium Signaling as a Therapeutic Target for Alzheimer’s Disease. Curr. Alzheimer Res. 2020, 17, 329–343. [Google Scholar] [CrossRef]
  12. Jadiya, P.; Kolmetzky, D.W.; Tomar, D.; Di Meco, A.; Lombardi, A.A.; Lambert, J.P.; Luongo, T.S.; Ludtmann, M.H.; Praticò, D.; Elrod, J.W. Impaired mitochondrial calcium efflux contributes to disease progression in models of Alzheimer’s disease. Nat. Commun. 2019, 10, 3885. [Google Scholar] [CrossRef] [Green Version]
  13. Godoy, J.A.; Rios, J.A.; Zolezzi, J.M.; Braidy, N.; Inestrosa, N.C. Signaling pathway cross talk in Alzheimer’s disease. Cell Commun. Signal. 2014, 12, 23. [Google Scholar] [CrossRef] [Green Version]
  14. Dhana, K.; Evans, D.A.; Rajan, K.B.; Bennett, D.A.; Morris, M.C. Healthy lifestyle and the risk of Alzheimer dementia: Findings from 2 longitudinal studies. Neurology 2020, 95, e374–e383. [Google Scholar] [CrossRef]
  15. Chakari-Khiavi, F.; Dolati, S.; Chakari-Khiavi, A.; Abbaszadeh, H.; Aghebati-Maleki, L.; Pourlak, T.; Mehdizadeh, A.; Yousefi, M. Prospects for the application of mesenchymal stem cells in Alzheimer’s disease treatment. Life Sci. 2019, 231, 116564. [Google Scholar] [CrossRef]
  16. Duncan, T.; Valenzuela, M. Alzheimer’s disease, dementia, and stem cell therapy. Stem Cell Res. Ther. 2017, 8, 111. [Google Scholar] [CrossRef]
  17. Lucke-Wold, B.P.; Logsdon, A.F.; Manoranjan, B.; Turner, R.C.; McConnell, E.; Vates, G.E.; Huber, J.D.; Rosen, C.L.; Simard, J.M. Aneurysmal Subarachnoid Hemorrhage and Neuroinflammation: A Comprehensive Review. Int. J. Mol. Sci. 2016, 17, 497. [Google Scholar] [CrossRef] [Green Version]
  18. Kim, J.; Lee, Y.; Lee, S.; Kim, K.; Song, M.; Lee, J. Mesenchymal Stem Cell Therapy and Alzheimer’s Disease: Current Status and Future Perspectives. J. Alzheimer’s Dis. 2020, 77, 1–14. [Google Scholar] [CrossRef]
  19. Boese, A.C.; Hamblin, M.H.; Lee, J.P. Neural stem cell therapy for neurovascular injury in Alzheimer’s disease. Exp. Neurol. 2020, 324, 113112. [Google Scholar] [CrossRef]
  20. Guo, M.; Yin, Z.; Chen, F.; Lei, P. Mesenchymal stem cell-derived exosome: A promising alternative in the therapy of Alzheimer’s disease. Alzheimer’s Res. Ther. 2020, 12, 109. [Google Scholar] [CrossRef]
  21. Hosseini, S.A.; Mohammadi, R.; Noruzi, S.; Mohamadi, Y.; Azizian, M.; Mousavy, S.M.; Ghasemi, F.; Hesari, A.; Sahebkar, A.; Salarinia, R.; et al. Stem cell- and gene-based therapies as potential candidates in Alzheimer’s therapy. J. Cell. Biochem. 2018, 119, 8723–8736. [Google Scholar] [CrossRef] [PubMed]
  22. Cosacak, M.I.; Bhattarai, P.; Kizil, C. Alzheimer’s disease, neural stem cells and neurogenesis: Cellular phase at single-cell level. Neural Regen. Res. 2020, 15, 824–827. [Google Scholar] [CrossRef] [PubMed]
  23. McGinley, L.M.; Kashlan, O.N.; Bruno, E.S.; Chen, K.S.; Hayes, J.M.; Kashlan, S.R.; Raykin, J.; Johe, K.; Murphy, G.G.; Feldman, E.L. Human neural stem cell transplantation improves cognition in a murine model of Alzheimer’s disease. Sci. Rep. 2018, 8, 14776. [Google Scholar] [CrossRef] [PubMed]
  24. Hayashi, Y.; Lin, H.T.; Lee, C.C.; Tsai, K.J. Effects of neural stem cell transplantation in Alzheimer’s disease models. J. Biomed. Sci. 2020, 27, 29. [Google Scholar] [CrossRef] [Green Version]
  25. Apodaca, L.A.; Baddour, A.A.D.; Garcia, C., Jr.; Alikhani, L.; Giedzinski, E.; Ru, N.; Agrawal, A.; Acharya, M.M.; Baulch, J.E. Human neural stem cell-derived extracellular vesicles mitigate hallmarks of Alzheimer’s disease. Alzheimer’s Res. Ther. 2021, 13, 57. [Google Scholar] [CrossRef] [PubMed]
  26. Revuelta, M.; Urrutia, J.; Villarroel, A.; Casis, O. Microglia-Mediated Inflammation and Neural Stem Cell Differentiation in Alzheimer’s Disease: Possible Therapeutic Role of K(V)1.3 Channel Blockade. Front. Cell. Neurosci. 2022, 16, 868842. [Google Scholar] [CrossRef]
  27. Reza-Zaldivar, E.E.; Hernández-Sapiéns, M.A.; Gutiérrez-Mercado, Y.K.; Sandoval-Ávila, S.; Gomez-Pinedo, U.; Márquez-Aguirre, A.L.; Vázquez-Méndez, E.; Padilla-Camberos, E.; Canales-Aguirre, A.A. Mesenchymal stem cell-derived exosomes promote neurogenesis and cognitive function recovery in a mouse model of Alzheimer’s disease. Neural Regen. Res. 2019, 14, 1626–1634. [Google Scholar] [CrossRef]
  28. Nakano, M.; Kubota, K.; Kobayashi, E.; Chikenji, T.S.; Saito, Y.; Konari, N.; Fujimiya, M. Bone marrow-derived mesenchymal stem cells improve cognitive impairment in an Alzheimer’s disease model by increasing the expression of microRNA-146a in hippocampus. Sci. Rep. 2020, 10, 10772. [Google Scholar] [CrossRef]
  29. Wei, H.; Xu, Y.; Chen, Q.; Chen, H.; Zhu, X.; Li, Y. Mesenchymal stem cell-derived exosomal miR-223 regulates neuronal cell apoptosis. Cell Death Dis. 2020, 11, 290. [Google Scholar] [CrossRef]
  30. Kim, H.J.; Cho, K.R.; Jang, H.; Lee, N.K.; Jung, Y.H.; Kim, J.P.; Lee, J.I.; Chang, J.W.; Park, S.; Kim, S.T.; et al. Intracerebroventricular injection of human umbilical cord blood mesenchymal stem cells in patients with Alzheimer’s disease dementia: A phase I clinical trial. Alzheimer’s Res. Ther. 2021, 13, 154. [Google Scholar] [CrossRef]
  31. Ubina, T.; Magallanes, M.; Srivastava, S.; Warden, C.D.; Yee, J.K.; Salvaterra, P.M. A Human Embryonic Stem Cell Model of Aβ-Dependent Chronic Progressive Neurodegeneration. Front. Neurosci. 2019, 13, 1007. [Google Scholar] [CrossRef] [Green Version]
  32. Fan, W.; Tang, S.; Fan, X.; Fang, Y.; Xu, X.; Li, L.; Xu, J.; Li, J.L.; Wang, Z.; Li, X. SIRT1 regulates sphingolipid metabolism and neural differentiation of mouse embryonic stem cells through c-Myc-SMPDL3B. eLife 2021, 10, e67452. [Google Scholar] [CrossRef]
  33. Kim, D.Y.; Choi, S.H.; Lee, J.S.; Kim, H.J.; Kim, H.N.; Lee, J.E.; Shin, J.Y.; Lee, P.H. Feasibility and Efficacy of Intra-Arterial Administration of Embryonic Stem Cell Derived-Mesenchymal Stem Cells in Animal Model of Alzheimer’s Disease. J. Alzheimer’s Dis. 2020, 76, 1281–1296. [Google Scholar] [CrossRef]
  34. Atkinson-Dell, R.; Mohamet, L. Induced Pluripotent Stem Cell-Derived Astroglia: A New Tool for Research towards the Treatment of Alzheimer’s Disease. Adv. Exp. Med. Biol. 2019, 1175, 383–405. [Google Scholar] [CrossRef]
  35. Butler Iii, R.R.; Kozlova, A.; Zhang, H.; Zhang, S.; Streit, M.; Sanders, A.R.; Laudanski, K.; Pang, Z.P.; Gejman, P.V.; Duan, J. The Genetic Relevance of Human Induced Pluripotent Stem Cell-Derived Microglia to Alzheimer’s Disease and Major Neuropsychiatric Disorders. Mol. Neuropsychiatry 2020, 5, 85–96. [Google Scholar] [CrossRef]
  36. Zhang, L.; Xu, M.; Ren, Q.; Liu, G.; Meng, S.; Xiahou, K.; Zhang, Y.; Jiang, N.; Zhou, W. Human Induced Pluripotent Stem Cell-Derived Neural Cells from Alzheimer’s Disease Patients Exhibited Different Susceptibility to Oxidative Stress. Stem Cells Dev. 2020, 29, 1444–1456. [Google Scholar] [CrossRef]
  37. Armijo, E.; Edwards, G.; Flores, A.; Vera, J.; Shahnawaz, M.; Moda, F.; Gonzalez, C.; Sanhueza, M.; Soto, C. Induced Pluripotent Stem Cell-Derived Neural Precursors Improve Memory, Synaptic and Pathological Abnormalities in a Mouse Model of Alzheimer’s Disease. Cells 2021, 10, 1802. [Google Scholar] [CrossRef]
  38. Aboul-Soud, M.A.M.; Alzahrani, A.J.; Mahmoud, A. Induced Pluripotent Stem Cells (iPSCs)-Roles in Regenerative Therapies, Disease Modelling and Drug Screening. Cells 2021, 10, 2319. [Google Scholar] [CrossRef]
  39. Kolagar, T.A.; Farzaneh, M.; Nikkar, N.; Khoshnam, S.E. Human Pluripotent Stem Cells in Neurodegenerative Diseases: Potentials, Advances and Limitations. Curr. Stem Cell Res. Ther. 2020, 15, 102–110. [Google Scholar] [CrossRef]
  40. Chang, C.Y.; Ting, H.C.; Liu, C.A.; Su, H.L.; Chiou, T.W.; Lin, S.Z.; Harn, H.J.; Ho, T.J. Induced Pluripotent Stem Cell (iPSC)-Based Neurodegenerative Disease Models for Phenotype Recapitulation and Drug Screening. Molecules 2020, 25, 2000. [Google Scholar] [CrossRef]
  41. Kwak, K.A.; Lee, S.P.; Yang, J.Y.; Park, Y.S. Current Perspectives regarding Stem Cell-Based Therapy for Alzheimer’s Disease. Stem Cells Int. 2018, 2018, 6392986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Yepes-Nuñez, J.; Urrutia, G.; Romero-Garcia, M.; Alonso-Fernandez, S. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Rev. Esp. Cardiol. (Engl. Ed.) 2021, 74, 790–799. [Google Scholar]
  43. Kim, J.Y.; Mo, H.; Kim, J.; Kim, J.W.; Nam, Y.; Rim, Y.A.; Ju, J.H. Mitigating Effect of Estrogen in Alzheimer’s Disease-Mimicking Cerebral Organoid. Front. Neurosci. 2022, 16, 816174. [Google Scholar] [CrossRef] [PubMed]
  44. Rajan, T.S.; Scionti, D.; Diomede, F.; Grassi, G.; Pollastro, F.; Piattelli, A.; Cocco, L.; Bramanti, P.; Mazzon, E.; Trubiani, O. Gingival Stromal Cells as an In Vitro Model: Cannabidiol Modulates Genes Linked with Amyotrophic Lateral Sclerosis. J. Cell. Biochem. 2017, 118, 819–828. [Google Scholar] [CrossRef]
  45. Campos, H.C.; Ribeiro, D.E.; Hashiguchi, D.; Hukuda, D.Y.; Gimenes, C.; Romariz, S.A.A.; Ye, Q.; Tang, Y.; Ulrich, H.; Longo, B.M. Distinct Effects of the Hippocampal Transplantation of Neural and Mesenchymal Stem Cells in a Transgenic Model of Alzheimer’s Disease. Stem Cell Rev. Rep. 2022, 18, 781–791. [Google Scholar] [CrossRef]
  46. Lin, Y.T.; Seo, J.; Gao, F.; Feldman, H.M.; Wen, H.L.; Penney, J.; Cam, H.P.; Gjoneska, E.; Raja, W.K.; Cheng, J.; et al. APOE4 Causes Widespread Molecular and Cellular Alterations Associated with Alzheimer’s Disease Phenotypes in Human iPSC-Derived Brain Cell Types. Neuron 2018, 98, 1141–1154.e7. [Google Scholar] [CrossRef] [Green Version]
  47. Zhao, J.; Fu, Y.; Yamazaki, Y.; Ren, Y.; Davis, M.D.; Liu, C.C.; Lu, W.; Wang, X.; Chen, K.; Cherukuri, Y.; et al. APOE4 exacerbates synapse loss and neurodegeneration in Alzheimer’s disease patient iPSC-derived cerebral organoids. Nat. Commun. 2020, 11, 5540. [Google Scholar] [CrossRef]
  48. Farahzadi, R.; Fathi, E.; Vietor, I. Mesenchymal Stem Cells Could Be Considered as a Candidate for Further Studies in Cell-Based Therapy of Alzheimer’s Disease via Targeting the Signaling Pathways. ACS Chem. Neurosci. 2020, 11, 1424–1435. [Google Scholar] [CrossRef]
  49. Xu, Z.; Nan, W.; Zhang, X.; Sun, Y.; Yang, J.; Lu, K.; Liu, Y.; Gao, Y.; Yang, F.; Mao, W.; et al. Umbilical Cord Mesenchymal Stem Cells Conditioned Medium Promotes Aβ25-35 phagocytosis by Modulating Autophagy and Aβ-Degrading Enzymes in BV2 Cells. J. Mol. Neurosci. 2018, 65, 222–233. [Google Scholar] [CrossRef]
  50. Chang, K.H.; Lee-Chen, G.J.; Huang, C.C.; Lin, J.L.; Chen, Y.J.; Wei, P.C.; Lo, Y.S.; Yao, C.F.; Kuo, M.W.; Chen, C.M. Modeling Alzheimer’s Disease by Induced Pluripotent Stem Cells Carrying APP D678H Mutation. Mol. Neurobiol. 2019, 56, 3972–3983. [Google Scholar] [CrossRef] [Green Version]
  51. Coronel, R.; Lachgar, M.; Bernabeu-Zornoza, A.; Palmer, C.; Domínguez-Alvaro, M.; Revilla, A.; Ocaña, I.; Fernández, A.; Martínez-Serrano, A.; Cano, E.; et al. Neuronal and Glial Differentiation of Human Neural Stem Cells Is Regulated by Amyloid Precursor Protein (APP) Levels. Mol. Neurobiol. 2019, 56, 1248–1261. [Google Scholar] [CrossRef]
  52. Lo Giudice, M.; Mihalik, B.; Turi, Z.; Dinnyés, A.; Kobolák, J. Calcilytic NPS 2143 Reduces Amyloid Secretion and Increases sAβPPα Release from PSEN1 Mutant iPSC-Derived Neurons. J. Alzheimer’s Dis. 2019, 72, 885–899. [Google Scholar] [CrossRef] [Green Version]
  53. Marzano, M.; Bejoy, J.; Cheerathodi, M.R.; Sun, L.; York, S.B.; Zhao, J.; Kanekiyo, T.; Bu, G.; Meckes, D.G., Jr.; Li, Y. Differential Effects of Extracellular Vesicles of Lineage-Specific Human Pluripotent Stem Cells on the Cellular Behaviors of Isogenic Cortical Spheroids. Cells 2019, 8, 993. [Google Scholar] [CrossRef] [Green Version]
  54. Meyer, K.; Feldman, H.M.; Lu, T.; Drake, D.; Lim, E.T.; Ling, K.H.; Bishop, N.A.; Pan, Y.; Seo, J.; Lin, Y.T.; et al. REST and Neural Gene Network Dysregulation in iPSC Models of Alzheimer’s Disease. Cell Rep. 2019, 26, 1112–1127.e9. [Google Scholar] [CrossRef] [Green Version]
  55. Van der Kant, R.; Langness, V.F.; Herrera, C.M.; Williams, D.A.; Fong, L.K.; Leestemaker, Y.; Steenvoorden, E.; Rynearson, K.D.; Brouwers, J.F.; Helms, J.B.; et al. Cholesterol Metabolism Is a Druggable Axis that Independently Regulates Tau and Amyloid-β in iPSC-Derived Alzheimer’s Disease Neurons. Cell Stem Cell 2019, 24, 363–375.e9. [Google Scholar] [CrossRef] [Green Version]
  56. Zhang, Z.; Sheng, H.; Liao, L.; Xu, C.; Zhang, A.; Yang, Y.; Zhao, L.; Duan, L.; Chen, H.; Zhang, B. Mesenchymal Stem Cell-Conditioned Medium Improves Mitochondrial Dysfunction and Suppresses Apoptosis in Okadaic Acid-Treated SH-SY5Y Cells by Extracellular Vesicle Mitochondrial Transfer. J. Alzheimer’s Dis. 2020, 78, 1161–1176. [Google Scholar] [CrossRef]
  57. Chen, Y.A.; Lu, C.H.; Ke, C.C.; Chiu, S.J.; Jeng, F.S.; Chang, C.W.; Yang, B.H.; Liu, R.S. Mesenchymal Stem Cell-Derived Exosomes Ameliorate Alzheimer’s Disease Pathology and Improve Cognitive Deficits. Biomedicines 2021, 9, 594. [Google Scholar] [CrossRef]
  58. Boutajangout, A.; Noorwali, A.; Atta, H.; Wisniewski, T. Human Umbilical Cord Stem Cell Xenografts Improve Cognitive Decline and Reduce the Amyloid Burden in a Mouse Model of Alzheimer’s Disease. Curr. Alzheimer Res. 2017, 14, 104–111. [Google Scholar] [CrossRef]
  59. Cui, Y.; Ma, S.; Zhang, C.; Cao, W.; Liu, M.; Li, D.; Lv, P.; Xing, Q.; Qu, R.; Yao, N.; et al. Human umbilical cord mesenchymal stem cells transplantation improves cognitive function in Alzheimer’s disease mice by decreasing oxidative stress and promoting hippocampal neurogenesis. Behav. Brain Res. 2017, 320, 291–301. [Google Scholar] [CrossRef]
  60. Harach, T.; Jammes, F.; Muller, C.; Duthilleul, N.; Cheatham, V.; Zufferey, V.; Cheatham, D.; Lukasheva, Y.A.; Lasser, T.; Bolmont, T. Administrations of human adult ischemia-tolerant mesenchymal stem cells and factors reduce amyloid beta pathology in a mouse model of Alzheimer’s disease. Neurobiol. Aging 2017, 51, 83–96. [Google Scholar] [CrossRef]
  61. Kim, H.S.; Lee, N.K.; Yoo, D.; Lee, J.; Choi, S.J.; Oh, W.; Chang, J.W.; Na, D.L. Lowering the concentration affects the migration and viability of intracerebroventricular-delivered human mesenchymal stem cells. Biochem. Biophys. Res. Commun. 2017, 493, 751–757. [Google Scholar] [CrossRef] [PubMed]
  62. Han, L.; Zhou, Y.; Zhang, R.; Wu, K.; Lu, Y.; Li, Y.; Duan, R.; Yao, Y.; Zhu, D.; Jia, Y. MicroRNA Let-7f-5p Promotes Bone Marrow Mesenchymal Stem Cells Survival by Targeting Caspase-3 in Alzheimer Disease Model. Front. Neurosci. 2018, 12, 333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Oh, S.; Son, M.; Choi, J.; Lee, S.; Byun, K. sRAGE prolonged stem cell survival and suppressed RAGE-related inflammatory cell and T lymphocyte accumulations in an Alzheimer’s disease model. Biochem. Biophys. Res. Commun. 2018, 495, 807–813. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, S.S.; Jia, J.; Wang, Z. Mesenchymal Stem Cell-Derived Extracellular Vesicles Suppresses iNOS Expression and Ameliorates Neural Impairment in Alzheimer’s Disease Mice. J. Alzheimer’s Dis. 2018, 61, 1005–1013. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, X.; Ma, S.; Yang, B.; Huang, T.; Meng, N.; Xu, L.; Xing, Q.; Zhang, Y.; Zhang, K.; Li, Q.; et al. Resveratrol promotes hUC-MSCs engraftment and neural repair in a mouse model of Alzheimer’s disease. Behav. Brain Res. 2018, 339, 297–304. [Google Scholar] [CrossRef] [Green Version]
  66. Wei, Y.; Xie, Z.; Bi, J.; Zhu, Z. Anti-inflammatory effects of bone marrow mesenchymal stem cells on mice with Alzheimer’s disease. Exp. Ther. Med. 2018, 16, 5015–5020. [Google Scholar] [CrossRef] [Green Version]
  67. Yu, S.; Hei, Y.; Liu, W. Upregulation of seladin-1 and nestin expression in bone marrow mesenchymal stem cell transplantation via the ERK1/2 and PI3K/Akt signaling pathways in an Alzheimer’s disease model. Oncol. Lett. 2018, 15, 7443–7449. [Google Scholar] [CrossRef]
  68. Esmaeilzade, B.; Artimani, T.; Amiri, I.; Najafi, R.; Shahidi, S.; Sabec, M.; Farzadinia, P.; Zare, M.; Zahiri, M.; Soleimani Asl, S. Dimethyloxalylglycine preconditioning enhances protective effects of bone marrow-derived mesenchymal stem cells in Aβ-induced Alzheimer disease. Physiol. Behav. 2019, 199, 265–272. [Google Scholar] [CrossRef]
  69. Hu, W.; Feng, Z.; Xu, J.; Jiang, Z.; Feng, M. Brain-derived neurotrophic factor modified human umbilical cord mesenchymal stem cells-derived cholinergic-like neurons improve spatial learning and memory ability in Alzheimer’s disease rats. Brain Res. 2019, 1710, 61–73. [Google Scholar] [CrossRef]
  70. Nasiri, E.; Alizadeh, A.; Roushandeh, A.M.; Gazor, R.; Hashemi-Firouzi, N.; Golipoor, Z. Melatonin-pretreated adipose-derived mesenchymal stem cells efficeintly improved learning, memory, and cognition in an animal model of Alzheimer’s disease. Metab. Brain Dis. 2019, 34, 1131–1143. [Google Scholar] [CrossRef]
  71. Eftekharzadeh, M.; Simorgh, S.; Doshmanziari, M.; Hassanzadeh, L.; Shariatpanahi, M. Human adipose-derived stem cells reduce receptor-interacting protein 1, receptor-interacting protein 3, and mixed lineage kinase domain-like pseudokinase as necroptotic markers in rat model of Alzheimer’s disease. Indian J. Pharmacol. 2020, 52, 392–401. [Google Scholar] [CrossRef]
  72. Guo, X.; Liu, Y.; Morgan, D.; Zhao, L.R. Reparative Effects of Stem Cell Factor and Granulocyte Colony-Stimulating Factor in Aged APP/PS1 Mice. Aging Dis. 2020, 11, 1423–1443. [Google Scholar] [CrossRef]
  73. Liu, Y.; Huber, C.C.; Wang, H. Disrupted blood-brain barrier in 5×FAD mouse model of Alzheimer’s disease can be mimicked and repaired in vitro with neural stem cell-derived exosomes. Biochem. Biophys. Res. Commun. 2020, 525, 192–196. [Google Scholar] [CrossRef]
  74. Mehrabadi, S.; Motevaseli, E.; Sadr, S.S.; Moradbeygi, K. Hypoxic-conditioned medium from adipose tissue mesenchymal stem cells improved neuroinflammation through alternation of toll like receptor (TLR) 2 and TLR4 expression in model of Alzheimer’s disease rats. Behav. Brain Res. 2020, 379, 112362. [Google Scholar] [CrossRef]
  75. Park, B.N.; Kim, J.H.; Lim, T.S.; Park, S.H.; Kim, T.G.; Yoon, B.S.; Son, K.S.; Yoon, J.K.; An, Y.S. Therapeutic effect of mesenchymal stem cells in an animal model of Alzheimer’s disease evaluated by β-amyloid positron emission tomography imaging. Aust. N. Z. J. Psychiatry 2020, 54, 883–891. [Google Scholar] [CrossRef]
  76. Park, D.; Choi, E.K.; Cho, T.H.; Joo, S.S.; Kim, Y.B. Human Neural Stem Cells Encoding ChAT Gene Restore Cognitive Function via Acetylcholine Synthesis, Aβ Elimination, and Neuroregeneration in APPswe/PS1dE9 Mice. Int. J. Mol. Sci. 2020, 21, 3958. [Google Scholar] [CrossRef]
  77. Ramezani, M.; Komaki, A.; Hashemi-Firouzi, N.; Mortezaee, K.; Faraji, N.; Golipoor, Z. Therapeutic effects of melatonin-treated bone marrow mesenchymal stem cells (BMSC) in a rat model of Alzheimer’s disease. J. Chem. Neuroanat. 2020, 108, 101804. [Google Scholar] [CrossRef]
  78. Zhao, J.; Su, M.; Lin, Y.; Liu, H.; He, Z.; Lai, L. Administration of Amyloid Precursor Protein Gene Deleted Mouse ESC-Derived Thymic Epithelial Progenitors Attenuates Alzheimer’s Pathology. Front. Immunol. 2020, 11, 1781. [Google Scholar] [CrossRef]
  79. Zhu, Q.; Zhang, N.; Hu, N.; Jiang, R.; Lu, H.; Xuan, A.; Long, D.; Chen, Y. Neural stem cell transplantation improves learning and memory by protecting cholinergic neurons and restoring synaptic impairment in an amyloid precursor protein/presenilin 1 transgenic mouse model of Alzheimer’s disease. Mol. Med. Rep. 2020, 21, 1172–1180. [Google Scholar] [CrossRef]
  80. Cone, A.S.; Yuan, X.; Sun, L.; Duke, L.C.; Vreones, M.P.; Carrier, A.N.; Kenyon, S.M.; Carver, S.R.; Benthem, S.D.; Stimmell, A.C.; et al. Mesenchymal stem cell-derived extracellular vesicles ameliorate Alzheimer’s disease-like phenotypes in a preclinical mouse model. Theranostics 2021, 11, 8129–8142. [Google Scholar] [CrossRef]
  81. Huang, D.; Cao, Y.; Yang, X.; Liu, Y.; Zhang, Y.; Li, C.; Chen, G.; Wang, Q. A Nanoformulation-Mediated Multifunctional Stem Cell Therapy with Improved β-Amyloid Clearance and Neural Regeneration for Alzheimer’s Disease. Adv. Mater. 2021, 33, 2006357. [Google Scholar] [CrossRef] [PubMed]
  82. Jeong, H.; Kim, O.J.; Oh, S.H.; Lee, S.; Reum Lee, H.A.; Lee, K.O.; Lee, B.Y.; Kim, N.K. Extracellular Vesicles Released from Neprilysin Gene-Modified Human Umbilical Cord-Derived Mesenchymal Stem Cell Enhance Therapeutic Effects in an Alzheimer’s Disease Animal Model. Stem Cells Int. 2021, 2021, 5548630. [Google Scholar] [CrossRef] [PubMed]
  83. Kuo, S.C.; Chio, C.C.; Yeh, C.H.; Ma, J.T.; Liu, W.P.; Lin, M.T.; Lin, K.C.; Chang, C.P. Mesenchymal stem cell-conditioned medium attenuates the retinal pathology in amyloid-β-induced rat model of Alzheimer’s disease: Underlying mechanisms. Aging Cell 2021, 20, e13340. [Google Scholar] [CrossRef] [PubMed]
  84. Lim, J.Y.; Park, S.I.; Park, S.A.; Jeon, J.H.; Jung, H.Y.; Yon, J.M.; Jeun, S.S.; Lim, H.K.; Kim, S.W. Potential application of human neural crest-derived nasal turbinate stem cells for the treatment of neuropathology and impaired cognition in models of Alzheimer’s disease. Stem Cell Res. Ther. 2021, 12, 402. [Google Scholar] [CrossRef] [PubMed]
  85. Lu, M.H.; Ji, W.L.; Chen, H.; Sun, Y.Y.; Zhao, X.Y.; Wang, F.; Shi, Y.; Hu, Y.N.; Liu, B.X.; Wu, J.W.; et al. Intranasal Transplantation of Human Neural Stem Cells Ameliorates Alzheimer’s Disease-Like Pathology in a Mouse Model. Front. Aging Neurosci. 2021, 13, 650103. [Google Scholar] [CrossRef]
  86. Neves, A.F.; Camargo, C.; Premer, C.; Hare, J.M.; Baumel, B.S.; Pinto, M. Intravenous administration of mesenchymal stem cells reduces Tau phosphorylation and inflammation in the 3xTg-AD mouse model of Alzheimer’s disease. Exp. Neurol. 2021, 341, 113706. [Google Scholar] [CrossRef]
  87. Santamaria, G.; Brandi, E.; Vitola, P.; Grandi, F.; Ferrara, G.; Pischiutta, F.; Vegliante, G.; Zanier, E.R.; Re, F.; Uccelli, A.; et al. Intranasal delivery of mesenchymal stem cell secretome repairs the brain of Alzheimer’s mice. Cell Death Differ. 2021, 28, 203–218. [Google Scholar] [CrossRef]
  88. Wang, H.; Liu, Y.; Li, J.; Wang, T.; Hei, Y.; Li, H.; Wang, X.; Wang, L.; Zhao, R.; Liu, W.; et al. Tail-vein injection of MSC-derived small extracellular vesicles facilitates the restoration of hippocampal neuronal morphology and function in APP / PS1 mice. Cell Death Discov. 2021, 7, 230. [Google Scholar] [CrossRef]
  89. Zhang, X.M.; Ouyang, Y.J.; Yu, B.Q.; Li, W.; Yu, M.Y.; Li, J.Y.; Jiao, Z.M.; Yang, D.; Li, N.; Shi, Y.; et al. Therapeutic potential of dental pulp stem cell transplantation in a rat model of Alzheimer’s disease. Neural Regen. Res. 2021, 16, 893–898. [Google Scholar] [CrossRef]
  90. Choi, J.M.; Park, H.S.; He, M.T.; Kim, Y.S.; Kim, H.Y.; Lee, A.Y.; Cho, E.J. Membrane-Free Stem Cells and Pyridoxal 5’-Phosphate Synergistically Enhance Cognitive Function in Alzheimer’s Disease Mouse Model. Antioxidants 2022, 11, 601. [Google Scholar] [CrossRef]
  91. Guo, W.; Zeng, Z.; Xing, C.; Zhang, J.; Bi, W.; Yang, J.; Shah, R.; Wang, D.; Li, Y.; Zhang, X.; et al. Stem cells from human exfoliated deciduous teeth affect mitochondria and reverse cognitive decline in a senescence-accelerated mouse prone 8 model. Cytotherapy 2022, 24, 59–71. [Google Scholar] [CrossRef]
  92. Liu, S.; Fan, M.; Xu, J.X.; Yang, L.J.; Qi, C.C.; Xia, Q.R.; Ge, J.F. Exosomes derived from bone-marrow mesenchymal stem cells alleviate cognitive decline in AD-like mice by improving BDNF-related neuropathology. J. Neuroinflam. 2022, 19, 35. [Google Scholar] [CrossRef]
  93. Wang, Y.; Jiang, J.; Fu, X.; Zhang, J.; Song, J.; Wang, Y.; Duan, L.; Shao, P.; Xu, X.; Zeng, L.; et al. Fe3O4@polydopamine nanoparticle-loaded human umbilical cord mesenchymal stem cells improve the cognitive function in Alzheimer’s disease mice by promoting hippocampal neurogenesis. Nanomedicine 2022, 40, 102507. [Google Scholar] [CrossRef]
  94. Zhang, H.A.; Yuan, C.X.; Liu, K.F.; Yang, Q.F.; Zhao, J.; Li, H.; Yang, Q.H.; Song, D.; Quan, Z.Z.; Qing, H. Neural stem cell transplantation alleviates functional cognitive deficits in a mouse model of tauopathy. Neural Regen Res. 2022, 17, 152–162. [Google Scholar] [CrossRef]
  95. Zhang, X.; Lei, T.; Wang, D.; Cai, S.; Hang, Z.; Yang, Y.; Bi, W.; Xiao, Z.; Du, H. Stem cells from human exfoliated deciduous teeth relieves Alzheimer’s disease symptoms in SAMP8 mice by up-regulating the PPARγ pathway. Biomed. Pharmacother. 2022, 152, 113169. [Google Scholar] [CrossRef]
  96. Wang, Y.; Jing, N.; Su, L.; Shi, C.; Zhang, P.; Wang, Z.; Sun, H.; Yang, J.; Liu, Y.; Wen, X.; et al. Establishment of induced pluripotent stem cell line (ZZUi009-A) from an Alzheimer’s disease patient carrying a PSEN1 gene mutation. Stem Cell Res. 2018, 27, 30–33. [Google Scholar] [CrossRef]
  97. Wang, Y.; Sun, H.; Yang, J.; Shi, C.; Liu, Y.; Xu, Y.; Zhang, J. Generation of induced pluripotent stem cell line (ZZUi0013-A) from a 65-year-old patient with a novel MEOX2 gene mutation in Alzheimer’s disease. Stem Cell Res. 2019, 34, 101366. [Google Scholar] [CrossRef]
  98. Dai, Q.; Zhang, W.; Guo, J.; Di, W.; Zhao, J.; Zhang, B.; Wang, Y. Generation of an induced pluripotent stem cell line (SIAISi003-A) from a 79-year-old patient with Alzheimer’s disease having APOE3/4 genetic background. Stem Cell Res. 2020, 48, 101949. [Google Scholar] [CrossRef]
  99. Wang, J.; Liu, X.; Du, X.; Ma, Z.; Liu, B.; Guo, R.; Feng, B.; Ma, J.; Cui, H. Induced pluripotent stem cells derived from one 70-years-old male donor with the APOE-ε4/ε4 alleles. Stem Cell Res. 2021, 53, 102395. [Google Scholar] [CrossRef]
  100. Wang, Z.; Sun, H.; Zhang, Q.; Yang, J.; Liu, Y.; Liu, H.; Shi, C.; Wang, Y.; Xu, Y. Generation of induced pluripotent stem cell line (ZZUi0024-A) from a 51-year-old patient with APP gene mutation in Alzheimer’ s disease. Stem Cell Res. 2021, 53, 102267. [Google Scholar] [CrossRef]
  101. Lee, Y.; Kim, M.; Lee, M.; So, S.; Kang, S.S.; Choi, J.; Kim, D.; Heo, H.; Lee, S.S.; Park, H.R.; et al. Mitochondrial genome mutations and neuronal dysfunction of induced pluripotent stem cells derived from patients with Alzheimer’s disease. Cell Prolif. 2022, 55, e13274. [Google Scholar] [CrossRef] [PubMed]
  102. Majd, S.; Power, J.; Majd, Z. Alzheimer’s Disease and Cancer: When Two Monsters Cannot Be Together. Front. Neurosci. 2019, 13, 155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Takagi, Y. History of Neural Stem Cell Research and Its Clinical Application. Neurol. Med.-Chir. 2016, 56, 110–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Stopa, E.G.; Gonzalez, A.M.; Chorsky, R.; Corona, R.J.; Alvarez, J.; Bird, E.D.; Baird, A. Basic fibroblast growth factor in Alzheimer’s disease. Biochem. Biophys. Res. Commun. 1990, 171, 690–696. [Google Scholar] [CrossRef] [PubMed]
  105. Yang, J.; Li, S.; He, X.B.; Cheng, C.; Le, W. Induced pluripotent stem cells in Alzheimer’s disease: Applications for disease modeling and cell-replacement therapy. Mol. Neurodegener. 2016, 11, 39. [Google Scholar] [CrossRef] [Green Version]
  106. Qin, C.; Lu, Y.; Wang, K.; Bai, L.; Shi, G.; Huang, Y.; Li, Y. Transplantation of bone marrow mesenchymal stem cells improves cognitive deficits and alleviates neuropathology in animal models of Alzheimer’s disease: A meta-analytic review on potential mechanisms. Transl. Neurodegener. 2020, 9, 20. [Google Scholar] [CrossRef]
  107. Volarevic, V.; Markovic, B.S.; Gazdic, M.; Volarevic, A.; Jovicic, N.; Arsenijevic, N.; Armstrong, L.; Djonov, V.; Lako, M.; Stojkovic, M. Ethical and Safety Issues of Stem Cell-Based Therapy. Int. J. Med. Sci. 2018, 15, 36–45. [Google Scholar] [CrossRef] [Green Version]
  108. Gorecka, J.; Kostiuk, V.; Fereydooni, A.; Gonzalez, L.; Luo, J.; Dash, B.; Isaji, T.; Ono, S.; Liu, S.; Lee, S.R.; et al. The potential and limitations of induced pluripotent stem cells to achieve wound healing. Stem Cell Res. Ther. 2019, 10, 87. [Google Scholar] [CrossRef] [Green Version]
  109. Wei, W.L.; Tsai, S.T.; Chiou, T.W.; Harn, H.J. Human-Induced Pluripotent Stem Cells and Herbal Small-Molecule Drugs for Treatment of Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 1327. [Google Scholar] [CrossRef] [Green Version]
  110. Patwardhan, A.G.; Belemkar, S. An update on Alzheimer’s disease: Immunotherapeutic agents, stem cell therapy and gene editing. Life Sci. 2021, 282, 119790. [Google Scholar] [CrossRef]
  111. Zhao, X.R.; Li, D.D.; Zhang, L.; Niu, Y.H.; Wang, W.Z.; Niu, B. Mesenchymal stem cell therapies for Alzheimer’s disease: Preclinical studies. Metab. Brain Dis. 2021, 36, 1687–1695. [Google Scholar] [CrossRef]
  112. Salwa; Kumar, L. Engrafted stem cell therapy for Alzheimer’s disease: A promising treatment strategy with clinical outcome. J. Control. Release 2021, 338, 837–857. [Google Scholar] [CrossRef]
Figure 1. Flow chart of the literature search following PRISMA 2020 guidelines. Searching databases were used as detailed in the main text. The reasons for exclusion of articles were as follows. Reason 1: stem cell was mentioned not as a direct therapeutic method, but as a medium for other medical treatments or preclinical research. Reason 2: the article included brief mentioning of Alzheimer’s disease, but was not targeted toward Alzheimer’s disease. Reason 3: Ineligible exposure group.
Figure 1. Flow chart of the literature search following PRISMA 2020 guidelines. Searching databases were used as detailed in the main text. The reasons for exclusion of articles were as follows. Reason 1: stem cell was mentioned not as a direct therapeutic method, but as a medium for other medical treatments or preclinical research. Reason 2: the article included brief mentioning of Alzheimer’s disease, but was not targeted toward Alzheimer’s disease. Reason 3: Ineligible exposure group.
Biomedicines 11 00120 g001
Figure 2. Distribution of stem cell types for AD therapies. (a) Cellular level studies, (b) Animal model studies, (c) Clinical level studies. Abbreviations: MSC (mesenchymal stem cell), NSC (neural stem cell), iPSC (induced pluripotent stem cell), ESC (embryonic stem cell), SHED (stem cell from human exfoliated deciduous teeth), SCF (stem cell factor), ATSC (adipose tissue mesenchymal stem cell), hADSC (human adipose-derived stem cell), hNTSC (human neural crest-derived nasal turbinate stem cell), MFSCE (membrane-free stem cell).
Figure 2. Distribution of stem cell types for AD therapies. (a) Cellular level studies, (b) Animal model studies, (c) Clinical level studies. Abbreviations: MSC (mesenchymal stem cell), NSC (neural stem cell), iPSC (induced pluripotent stem cell), ESC (embryonic stem cell), SHED (stem cell from human exfoliated deciduous teeth), SCF (stem cell factor), ATSC (adipose tissue mesenchymal stem cell), hADSC (human adipose-derived stem cell), hNTSC (human neural crest-derived nasal turbinate stem cell), MFSCE (membrane-free stem cell).
Biomedicines 11 00120 g002
Figure 3. Collection of pathological effects for different types of stem cells. Abbreviations: MSC (mesenchymal stem cell), NSC (neural stem cell), iPSC (induced pluripotent stem cell), ESC (embryonic stem cell), SHED (stem cell from human exfoliated deciduous teeth), SCF (stem cell factor), ATSC (adipose tissue mesenchymal stem cell), hADSC (human adipose-derived stem cell), hNTSC (human neural crest-derived nasal turbinate stem cell), MFSCE (membrane-free stem cell).
Figure 3. Collection of pathological effects for different types of stem cells. Abbreviations: MSC (mesenchymal stem cell), NSC (neural stem cell), iPSC (induced pluripotent stem cell), ESC (embryonic stem cell), SHED (stem cell from human exfoliated deciduous teeth), SCF (stem cell factor), ATSC (adipose tissue mesenchymal stem cell), hADSC (human adipose-derived stem cell), hNTSC (human neural crest-derived nasal turbinate stem cell), MFSCE (membrane-free stem cell).
Biomedicines 11 00120 g003
Table 1. Summary of cellular level studies on stem cell therapy for AD (2017–2022).
Table 1. Summary of cellular level studies on stem cell therapy for AD (2017–2022).
AuthorYearType of Stem CellInterventionControl GroupMeasured Outcomes of InterestReference
Animal-induced stem cellsFarahzadi et al.2020BM-MSCInjection of BM-MSCs with Aβ-treated neural cellsAβ-treated neural cells without BM-MSC injectionSignificant increase in telomerase activity, which indicates mitigation of AD[48]
Human-oriented stem cellsXu et al.2018ucMSCCulture ucMSCs to BV2 cell line with Aβ25–35 oligomersBV2 cell line without culturing ucMSCsMSCs inhibited proliferation of BV2 cells, mediated microglial autophagy, and enhanced clearance of Aβ, hence mitigating AD[49]
Lin et al.2018iPSCUsing CRISPR/Cas9 to generate isogenic iPSCs harboring homozygous APOE4 alleles from unaffected parental APOE3 cellsParental APOE3 cells without injection of isogenic iPSC linesAPOE4 iPSC-derived neurons recapitulate phenotypes of AD at multiple levels, including increased synapse number and elevated Aβ42 secretion[46]
Chang et al.2019iPSCInjection of a potential Aβ aggregation reducer indole compound NC009–1 with iPSC on live human neurons from AD patientsAD neurons without injection of iPSCImproved neurite outgrowth in AD-iPSC-derived neurons[50]
Coronel et al.2019hNSCTransiently increase APP level in hNSCs hNSCs without increasing APP level Promoted gliogenesis and inhibited neurogenesis in hNSCs[51]
Giudice et al.2019iPSCInjection of γ-secretase inhibitor to iPSC-derived neurons from AD patientsAD neurons without injection of iPSC-derived neuronsIncreased expression of CaSR protein and modulated PSEN1 expression in plasma membrane of AD neurons[52]
Marzano et al.2019hiPSCInjection of hiPSC-derived extracellular vesicles (EVs) to AD-associated SY-UBH cell linesSY-UBH cells without hiPSC injectionhiPSC-derived EVs exhibited neural protective abilities in Aβ42 oligomer-treated cultures, enhancing cell viability and reducing oxidative stress[53]
Meyer et al.2019iPSCiPSCs generated from sporadic AD (SAD) patientsiPSCs generated from age-matched controls of familial AD (FAD) patientsSAD iPSC lines showed a significant level of increase in neural differentiation-related gene expression, leading to premature neuronal differentiation, and reduced neural progenitor cell renewal[54]
van der Kant et al.2019iPSCiPSC-derived neurons from SAD patientsiPSC-derived neurons from FAD patientsAllosteric activation of CYP46A1 lowered cholesteryl esters (CE) in iPSC-derived neurons, indicating a druggable CYP46A1-CE-Tau axis in AD.[55]
Zhang et al. 2020hucMSCTreatment of hucMSC conditioned medium to AD cellular model established by okadaic acid-treated SH-SY5Y cellsSH-SY5Y cells without treatment of hucMSChucMSCs significantly decreased tau phosphorylation at Thur181 level, and alleviated intracellular and mitochondrial oxidative stress in AD cells[56]
Zhao et al.2020iPSCCerebral organoid model using iPSCs with APOE3 or APOE4 genotype from individuals with AD dementiaCerebral organoid model using iPSCs with APOE3 or APOE4 genotype from individuals with normal cognitionCerebral organoids model using iPSCs from AD patients carrying APOE4 show greater apoptosis and decreased synaptic integrity[47]
Chen et al.2021MSCCulture MSC-exosomes into human neural cell with AD mutationsHuman neural cells without culturing MSC-exosomesImprovement in brain glucose metabolism and cognitive functioning[57]
Table 2. Summary of animal model studies on stem cell therapy for AD in chronological order (2017–2022).
Table 2. Summary of animal model studies on stem cell therapy for AD in chronological order (2017–2022).
AuthorYearType of Stem CellInterventionControl GroupMeasured Outcomes of InterestReference
Boutajangout et al.2017hucMSCDirect injection of hucMSCs into carotid artery of APP/PS1 Tg AD model miceAD mice without injection of hucMSCsReduction of amyloid beta burden in cortex and hippocampus which correlated with a reduction of cognitive loss[58]
Cui et al.2017ucMSCIntravenous transplantation of ucMSCs to Tg2576 mice, which express AD-like APP pathological formsTg2576 mice without transplantation of ucMSCsReduced oxidative stress in hippocampus of AD mice due to decrease of malondialdehyde (MDA) and hence up-regulated neuronal synaptic plasticity[59]
Harach et al.2017MSCAdminister MSC intravenously to APP/PS1 transgenic mice that developed cerebral AβAPPPS1 transgenic mice without administering MSCsReduced soluble cerebral Aβ levels and increased Aβ-degrading enzymes to modulate cerebral cytokines[60]
Kim et al.2017hUCB-MSCInjection of hUCB-MSCs in C3H/C57 wild-type miceC3H/C57 mice without injection of hUCB-MSCIncreased cell viability and rate of cell migration in the brain intracerebroventricular route, hence mitigating AD.[61]
Han et al.2018BM-MSCExposure to MicroRNA let-7f-5p modified MSCs in vitro in C57BL/6 miceC57BL/6 mice without MSC transplantationIncreased caspase-3 expression and hence decreased cytotoxicity for AD models[62]
McGinley et al.2018hNSCTransplantation of hNSC targeted to fimbria fornix of APP/PS1 murineAPP/PS1 mice without transplantation of hNSCsReduced amyloid plaque load and increased recruitment of activated microglia, indicating functional and pathological improvements in AD mice[23]
Oh et al. 2018sRAGE-MSCInjection of sRAGE-MSCs with Aβ1–42 to entorhinal cortices of male Sprague Dawley ratsSprague Daley rats without injection of sRAGE-MSCsLonger survival time for mice with sRAGE-MSC injection due to prevented apoptosis, and decrease in neuron numbers[63]
Wang et al.2018MSCIntracerebroventricular injection of MSC-derived extracellular vesicles to APP/PS1 miceAPP/PS1 mice without injection of MSCsAlleviated exogenous Aβ-induced iNOS mRNA and protein expression, therefore improved cognitive behavior and rescued impairment of CA1 synaptic transition and long-term potentiation in AD mice[64]
Wang et al.2018ucMSCInjection of ucMSCs with resveratrol to hippocampus of AD miceAD mice without ucMSC injectionImproved learning and memory, enhanced neurogenesis, and alleviated neural apoptosis in AD mice[65]
Wei et al. 2018BM-MSCInjection of BM-MSCs on APP/PS1 miceAPP/PS1 mice without BM-MSC injectionReduced Aβ1–42 content and BACE1 gene expression; ameliorated symptoms of AD [66]
Yu et al.2018BM-MSCTransplantation of BM-MSCs to murine models with ADAD murine model without BM-MSC transplantationIncreased protein expression of seladin-1 and nestin, and hence reduced neurodegeneration[67]
Esmaeilzade et al.2019MSCIncubation of MSCs with dimethyloxalylglycine (DMOG) in Aβ-injected ratsAβ-injected rats without MSC incubationIncreased cell viability, migration, and expression of CXCR4, CCR2, Nrf2, and HIF-1α, which enhanced antioxidant capacity in the hippocampus[68]
Hu et al.2019ucMSCTransplantation of ucMSCs with brain-derived neurotrophic factor (BDNF) to mice with Aβ1–42AD mice without transplantation of ucMSCsSignificantly improved spatial learning and memory abilities in the AD rats; increased release of acetylcholine and ChAT expression in the hippocampus[69]
Nasiri et al.2019AD-MSCInjection of melatonin-pretreated AD-MSCs to male Wistar ratsMale Wistar rats without injection of AD-MSCsCleared Aβ deposition and reduced microglial cells[70]
Reza-Zaldivar et al.2019MSCAdministration of MSC-derived exosomes to amyloid 1–42 treated AD miceAD mice without injection of MSC-derived exosomesAlleviated beta amyloid 1–42-induced cognitive impairment[27]
Eftekharzadeh et al.2020hADSCIntravenous injection of hADSCs to murine model of ADMurine model without administration of hADSCshADSCs significantly decreased the expression of necroptotic markers and reduced necroptosis and declined death of neuronal cells in the hippocampus of AD rats[71]
Guo et al.2020SCFSubcutaneously injection of SCF to APP/PS1 transgenic mice with C57BL/6J genetic backgroundAPP/PS1 transgenic mice without injection of SCFIncreased association of TREM2+/Iba1+ cells with Aβ plaques and enhanced cerebral expression that ameliorated AD pathology at late stage[72]
Kim et al.2020ES-MSCIntra-arterial administration of ES-MSCs in AD rat modelAD rats without administration of ES-MSCsSignificantly inhibited Aβ-induced cell death in hippocampus and promoted autophagolysosomal clearance of Aβ[33]
Liu et al.2020NSC Injection of NSC-derived exosomes to 5 × FAD mice5 × FAD mice without injection of NSC-derived exosomesReversed AD-caused blood–brain barrier deficiency[73]
Mehrabadi et al.2020ATSCsInjection of hypoxic-conditioned medium from ATSCs intravenously to AD miceAD mice without injection of ATSCsDecreased beta amyloid plaques, TLR2 and TLR4 expression and enhanced neuronal survival[74]
Park et al.2020BM-MSCIntravenous injection of BM-MSCs to 3xTg AD miceAD mice without injection of BM-MSCsEnhanced memory function and less β-amyloid-immunopositive plaques[75]
Park et al.2020NSCTransplantation of NSCs into APPswe/PS1dE9 AD model mice AD mice without NSC transplantationIncreased ACh (acetylcholinesterase) level and improved learning and memory function[76]
Ramezani et al.2020BM-MSCTransplantation of BM-MSCs to male Wistar ratsWistar rats without BM-MSC transplantationEnhanced learning, cognition and memory that mitigated neurodegeneration of AD[77]
Zhao et al.2020ESCTransplantation of mouse ESC-derived thymic epithelial progenitors into AD miceWithout ESC transplantationReduced cerebral Aβ plaque load and improved cognitive performance with increased T cell number[78]
Zhu et al.2020NSCImplantation of NSCs to hippocampus of APP/PS1 Tg (transgenic) AD miceAD mice without NSC implantationProtected cholinergic neurons, restored synaptic impairment in amyloid precursor and hence improved learning and memory function[79]
Apodaca et al.2021hNSCIntravenous injection of hNSC-derived extracellular vesicles on 5xFAD accelerated transgenic AD miceAD mice without injection of hNSC-derived extracellular vesiclesReduced dense core Aβ plaque accumulation and microglial activation, restoration of homeostatic levels of circulating pro-inflammatory cytokines in AD mice, with improved cognition and synaptic function[25]
Armijo et al.2021iPSCStereotaxically injection of iPSC-derived neural precursors to hippocampus of 3xTg AD miceAD mice without injection of iPSC-derived neural precursorsImproved memory, synaptic plasticity, and reduced brain pathology, including a reduction of amyloid and tangles deposits[37]
Cone et al.2021MSCIntranasally injection of NSC derived extracellular vesicles to non-transgenic 5xFAD miceAD mice without NSC injectionLowered Aβ plaque load in the hippocampus. Less colocalization between GFAP and Aβ plaques and hence better cognition functions[80]
Huang et al.2021Nanoformulation-mediated NSCInjection of nanoformulation-mediated NSC into APPswe/PS1dE9 double transgenic mouse modelMouse model without NSC injectionImproved neural regeneration, and efficient and long-lasting Aβ degradation[81]
Jeong et al.2021hucMSCIntravenous transplantation of hucMSCs into Aβ injected AD animal modelAD animal model without hucMSC transplantationSuperior neurogenesis and anti-inflammation properties with increased NEP in hippocampus[82]
Kuo et al.2021MSCIntracerebroventricular administration of MSC-conditioned medium to Aβ-induced rat modelRat model without administration of MSCDecreased expression of tight junction proteins, SIRT1 and β-catenin, which attenuated retinal pathology of AD[83]
Lim et al.2021hNTSCTransplantation of hNTSC into 5xFAD transgenic AD miceAD mice without hNTSC transplantationReduced Aβ42 levels and plaque formation in the brain, increased survival of hippocampal and cortex neurons[84]
Lu et al.2021hNSCIntranasal transplantation of hNSCs into APP/PS1 transgenic AD miceAD mice without hNSC transplantationAttenuated beta-amyloid accumulation by upregulating the expression of beta-amyloid degrading enzymes, insulin degrading enzymes and neprilysin, which ameliorated neuroinflammation, cholingergic dysfunction, and synaptic loss[85]
Neves et al.2021BM-MSCAdminister of allogeneic BM-MSCs intravenously in 3xTg AD miceAD mice without administering BM-MSCsReduced β-secretase cleavage of amyloid precursor protein and decreased tau phosphorylation[86]
Santamaria et al.2021MSCIn vivo systematic administration of MSCs to APP/PS1 AD miceAD mice without administration of MSCsInduced persistent memory recovery and reduced plaques with β-amyloid oligomers[87]
Wang et al.2021MSCTail-vein injection of MSC-derived small extracellular vesicles into APP/PS1 AD miceAD mice without injection of MSCsRestored hippocampal neuronal morphology, with improved cognitive impairments and reduced neuronal loss[88]
Zhang et al.2021Dental pulp stem cellInjection of 5 × 10 dental pulp stem cells into the hippocampus of AD miceAD mice without injection of dental pulp stem cellsIncreased expression of neuron-related doublecortin, NeuN, and neurofilament 200 in the hippocampus with decreased expression of Aβ, hence improving cognitive and behavioral abilities[89]
Choi et al.2022MFSCETreatment of MFSCE on Aβ25–35-injected AD miceAD mice without MFSCE treatmentSuppressed Bax and cleaved caspase-3 protein expression, downregulated amyloidogenic-pathway-related protein expressions and hence improved cognitive functioning[90]
Guo et al.2022SHEDSHEDs cultured in vitro and injected into AD SAMP8 mice by caudal veinAD mice without SHED injectionImproved cognitive ability and reversed memory loss through the recovery of dysfunctional mitochondria[91]
Liu et al.2022BM-MSCLateral ventricle administration of BM-MSCs to adult C57BL/6 AD miceAD mice without administration of BM-MSCsInhibited hyper activation of microglia and astrocytes in the hippocampus of AD mice[92]
Wang et al.2022hucMSCInjection of hucMSCs with Fe3O4 polydopamine nanoparticles into APP/PS1 transgenic miceAD mice without hucMSC injectionImproved memory and cognitive ability of AD by increased expression of brain-derived neurotrophic factor[93]
Zhang et al.2022NSCTransplantation of NSCs into hippocampal CA1 region of rTg (tau P301L) 4510 mouse modelMouse model without transplantation of NSCsReduced abnormal aggregation of tau, and hence improvements in short-term memory[94]
Zhang et al.2022SHEDInjection of SHED into SAMP8 AD miceAD mice without SHED injectionRelieved AD symptoms by up-regulating PPARγ pathway[95]
Table 3. Summary of clinical level studies on stem cell therapy for AD in chronological order (2017–2022).
Table 3. Summary of clinical level studies on stem cell therapy for AD in chronological order (2017–2022).
AuthorYearType of Stem CellInterventionControl GroupMeasured Outcomes of InterestReference
Wang et al.2018iPSCInduction of iPSC with Sendai-virus delivery systemN/AObtain iPSC cell line (ZZUi009-A) from AD patient with PSEN1 gene mutation[96]
Wang et al.2019iPSCInduction of iPSC with novel MEOX2 mutation in a family with ADN/AObtain iPSC cell line (ZZUi0013-A) with episomal plasmids expressing OCT3/4, SOX2, KLF4, LIN28, and L-MYC genes[97]
Dai et al.2020iPSCGeneration of iPSC from peripheral blood mononuclear cells with AD patient with APOE3/4 genotypeN/AWith immunocytochemistry iPSC displayed potential to differentiate spontaneously into three germ layers in vitro[98]
Kim et al.2021hUCB-MSCIntracerebroventricular injection of hUCB-MSCs to AD patientsAD patients without hUCB-MSC injectionPhase I clinical trial showed significant effect in mitigating neurodegeneration despite few adverse events (fever, headache, vomit)[30]
Wang et al.2021iPSCInduction of iPSC from male AD patient with APOE-ε4/ε4 allelesN/AObtain iPSCs with differentiation potential for treating neurological disorder and multiple sclerosis[99]
Wang et al.2021iPSCInduction of iPSC with APP gene mutation in a female patient with ADN/AObtain iPSC cell line (ZZUi0024-A) with dermal fibroblasts expressing OCT3/4, SOX2, KLF4, LIN28, and L-MYC genes[100]
Lee et al.2022iPSCInduction of iPSC from AD patients with mtDNA mutationsN/AmtDNA mutations induced growth advantage with higher viability and proliferation, lower mitochondrial respiration, and membrane potential[101]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Duan, Y.; Lyu, L.; Zhan, S. Stem Cell Therapy for Alzheimer’s Disease: A Scoping Review for 2017–2022. Biomedicines 2023, 11, 120. https://doi.org/10.3390/biomedicines11010120

AMA Style

Duan Y, Lyu L, Zhan S. Stem Cell Therapy for Alzheimer’s Disease: A Scoping Review for 2017–2022. Biomedicines. 2023; 11(1):120. https://doi.org/10.3390/biomedicines11010120

Chicago/Turabian Style

Duan, Yunxiao, Linshuoshuo Lyu, and Siyan Zhan. 2023. "Stem Cell Therapy for Alzheimer’s Disease: A Scoping Review for 2017–2022" Biomedicines 11, no. 1: 120. https://doi.org/10.3390/biomedicines11010120

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop