Open access peer-reviewed chapter

New Prospects for Stem Cell Therapy in Alzheimer’s Disease

Written By

Kun Jiang, Yongqi Zhu and Lei Zhang

Submitted: 03 August 2021 Reviewed: 07 September 2021 Published: 24 November 2021

DOI: 10.5772/intechopen.100334

From the Edited Volume

Hippocampus - Cytoarchitecture and Diseases

Edited by Xinhua Zhang

Chapter metrics overview

303 Chapter Downloads

View Full Metrics

Abstract

Alzheimer’s disease (AD) is a kind of neurodegenerative disease with insidious onset and progressive progression. The etiology of AD may be related to the loss of neurons, astrocytes, and microglial in the nervous system. Exogenous stem cell transplantation has brought hope to the treatment of AD. Stem cell transplantation can reduce amyloid β-protein (Aβ) deposition and Tau phosphorylation, and provide secretory factor support to improve learning and memory deficits. The purpose of this review is to provide an overview of the relationship between different stem cell species and the treatment of AD, and also summarize current experimental stem cell therapy strategies and their potential clinical applications in the future.

Keywords

  • Stem cells
  • Therapy
  • Alzheimer’s disease (AD)

1. Introduction

According to the World Alzheimer Report 2019, more than 50 million people worldwide suffer from dementia. It is expected to grow to 152 million by 2050. The current cost of treating dementia is $1 trillion a year, and that cost is expected to double by 2030. There are more than 200 subtypes of dementia, of which 50 to 60 percent are caused by Alzheimer’s disease (AD). The concept of the disease was proposed by Alois Alzheimer in 1907. It was later recognized as the most common neurodegenerative disease. Although decades have passed since the discovery of the pathological mechanism of Alzheimer’s disease, we still do not know what causes the disease. It is well known that Alzheimer’s disease is a sporadic, age-related disease, with only a small proportion caused by genetic factors. The disease is characterized by a progressive decline in cognitive function. Clinically, these patients present with short-term memory impairments that interfere with activities of daily living, followed by impairments in other cognitive areas such as language, logical understanding, orientation, executive function, judgment, behavior, and finally motor impairments [1]. The pathological features of AD include: Senile Plaques (SP) formed by the deposition of amyloid β-protein (Aβ) outside neurocyte; The abnormal phosphorylation of intracellular Tau protein results in the neurofibrillary tangles (NFTs); Synaptic loss, neuroinflammation, neurocyte apoptosis in the neocortex and hippocampus of the brain. The pathological manifestations were brain atrophy [2, 3, 4, 5]. In this review, we believe that the most effective strategies should target the biological feature which is most associated with symptoms, the loss of synapses, to treat the disease. Specifically, we focus on recent advances in cell-based therapies that aim at repopulation or regeneration of degenerating neuronal networks in AD [6].

Advertisement

2. Alzheimer’s Disease’s neuropathology

As mentioned in the background, we group the pathological changes of Alzheimer’s disease into two types, which provide evidence of the disease’s occurrence and progression: (1) Positive lesions. The main findings include SP caused by Aβ deposition and NFTs caused by abnormal phosphorylation of intracellular Tau protein. Otherwise, dystrophic neurites, neuropil threads and various other sediments found in the brains of patients with AD also falls into this category. (2) Negative lesions, which can also called loss type lesions. The main clinical manifestation is brain atrophy due to loss of synapses. At the same time, other factors, including neuroinflammation, oxidative stress, and damage to cholinergic neurons, are all important factors leading to the occurrence of neurodegenerative diseases.

2.1 Senile plaques (SP)

The SP are extracellular deposits of Aβ with different morphological forms, including neuritic, diffuse, dense-cored, or classic and compact type plaques [5].

The formation of Aβ is from the amyloidogenic cleavage of human amyloid precursor protein (APP) [7]. The anomalous processing of APP by β-secretases and γ-secretases leads to production of Aβ40 and Aβ42 monomers, which further oligomerize and aggregate into SP [8, 9]. Although soluble Aβ40 is much more abundant than soluble Aβ42, Aβ42 exhibits a higher propensity for aggregation, due to hydrophobicity within its two terminal residues. Indeed, Aβ42 is the main component of amyloid plaques and is shown to be neurotoxic [10]. Recent neuroimaging and neuropathology researches reveal that Aβ sedimentation is mainly related to cognitive disorder of the old, and it is not very relevant with other clinical features [11].

2.2 Neurofibrillary tangles (NFTs)

Tau protein is mainly distributed in neurons. Repeated Pro-Gly-Gly-Gly fragments help it bind to tubulin and maintain the structural stability of microtubules. The presence of Tau protein contributes to the maintenance of cytoskeleton and the integrity of axon transport [12]. NFTs are filamentous structures filled in the cytoplasm of neurons -- paired helical fibers (PHF). The reason of Tau hyperphosphorylation is the increased protein kinase activity. Protein kinase activity such as glycogen synthase kinase 3β (GSK-3β) activity can be decreased to reduce phosphorylation. Meanwhile, decreased phosphatase activity is also the reason of hyperphosphorylation. In addition, the lack of glucose in the brain can make Tau hyperphosphorylated by mediating the signal pathway of p38 mitogen-activated protein kinase (MAPK). Increasing the level of glucose in the brain may provide a new idea for treating AD, by using a pharmacological model of glucose deprivation and investigated its effect on Tau phosphorylation, synaptic function and cognition in a relevant transgenic mouse model of tauopathy, the h-Tau mouse [13]. It has been shown that phosphorylation of Tau protein at the early stage of AD inhibits Aβ toxicity, being that Tau phosphorylation-mediated by p38 MAPK can antagonize the postsynaptic excitation toxicity caused by Aβ [14, 15].

2.3 Synaptic loss

Soluble Aβ collaborate with pTau to induce synapse loss and cognitive impairment in AD [16]. Metabolism of Aβ and Tau proteins is crucially influenced by autophagy. Autophagy is a lysosome-dependent, homeostatic process, in which organelles and proteins are degraded and recycled into energy [17]. Neuroplasticity is an ongoing process that responds to the activity, injury, and death of neurons, including the regulation of the structure and function of axons, dendrites and synapses [18]. Overdeposition of Aβ and abnormal phosphorylation of Tau both lead to decreased neuroplasticity, which is manifested in a series of clinical symptoms caused by synaptic loss in AD [12, 19]. Aβ and Tau both trigger mitochondrial alterations. Some evidence suggests that mitochondrial perturbation acts as a key factor that is involved in synaptic failure and degeneration in AD [20]. Synaptic plasticity and long-term potentiation (LTP) are all about N-methyl-D-aspartate receptor (NMDAR). Aβ oligomer facilitates astrocytes (AS) to release glutamate by a7nAChR and activates NMDAR, making extracellular regulated protein kinases (ERK) signaling pathway to be suppressed and finally suppressing LTP, therefore the synaptic damages caused by NMDAR hyperactivation are the possible mechanisms of AD occurring [21].

2.4 Neuroinflammation

The AD pathophysiology entails chronic inflammation involving innate immune cells including microglia, astrocytes, and other peripheral blood cells. Inflammatory mediators such as cytokines and complements are also linked to AD pathogenesis [22, 23]. Activation of microglia can induce the production of inflammasomes, which in turn increase inflammatory cytokines, and may eventually result in Aβ deposition [24, 25]. Studies have shown that after being activated, astrocytes will release the corresponding cytokines, which can lead to the enhancement of neuronal toxicity, as well as a decreased outgrowth of neuronal processes and an overall decreased activity rate [26]. Recent studies have shown that there is a direct interaction between microglia and astrocytes. In the form that once microglia are activated, they can lead to activation of astrocytes, thus forming feed-forward loops that are harmful to the surrounding environment [26]. The mechanism showed that when being activated, microglia release IL-1α, TNFα and C1q and astrocytes become activated. Microglia and astrocytes are major modulators of inflammation in the brain, and they are also the major sources of apolipoprotein E (ApoE) in the brain. ApoE is a multifunctional protein with central roles in lipid metabolism. It transports lipids, including cholesterol, through the cerebrospinal fluid (CSF) and plasma [27, 28]. Earlier studies have shown that the presence of ApoE helps to inhibit glial activation of lipopolysaccharides in glial cell culture experiments, suggesting that ApoE may exert a protective anti-inflammatory effect [29, 30]. Moreover, the exacerbated proinflammatory state that occurs during this period of AD can trigger the hyperphosphorylation of Tau. Several of the kinases responsible for Tau phosphorylation are activated by proinflammatory mediators and have been shown to worsen Tau pathology [31].

2.5 Cholinergic neurons’ injuries

The Acetylcholine (ACh) receptor (AChR) is a vital membrane protein on which ACh acts as a neurotransmitter. The cholinergic receptors are broadly categorized as muscarinic ACh receptors (mAChR) and nicotinic ACh receptors (nAChR) on the basis of their exogenous agonists [32]. ACh plays an important role in human memory function and is strongly associated with age-related dementia such as AD, in which hippocampal dependent learning dysfunction is prominent. Cholinergic neurons densely dominate the hippocampus and mediate the production of episodic and semantic memory [33]. In patients with AD, the synthesis, release and uptake of ACh in the hippocampus, neocortex and cerebrospinal fluid were decreased, the choline acetyltransferase (AChE) was significantly decreased, and the activity of acetylcholinesterase was decreased [34]. Clinically, the main method of drug treatment for AD is to improve the function of the brain’s cholinergic system. Although inhibitors of acetylcholinesterase is a symptomatic relief treatment with marginal benefits, it is currently the most available clinical treatment which gives desperate AD patients a glimmer of hope [35].

Advertisement

3. Stem cell therapy for AD

There are some theoretical approaches to treat early AD. One is to target upregulation of resident neural stem cells (NSCs) niches within the adult brain. In fact, this regulation is to stimulate the development of adult hippocampal nerve, which has reached the purpose of compensating the degenerated nerve. Adult hippocampal neurogenesis may play a key role in learning and memory, so promoting this endogenous process may help improve amnesia in patients with early AD. Another approach is to up-regulate growth factors that are known to modulate neurogenesis integrally, either through drug therapy or gene therapy or, as we describe in this paper, through stem cell therapy. This type of growth factor includes brain-derived neurotrophic factor (BDNF) [36, 37], insulin growth factor-1 (IGF-1) [38], nerve growth factor (NGF) [39, 40, 41, 42], vascular endothelial growth factor (VEGF) [43, 44] and so on. Stem cell therapy aims to rescue cognitive function by introducing exogenous stem cells to restore degenerated neural networks. These stem cells can be used as cell delivery systems through the natural or induced production of neuroprotective growth factors utilizing the paracrine “bystander” mechanism. Alternatively, therapeutic recovery may occur through the differentiation and involvement of stem cells in refilling degenerated neuronal circuits. It’s a finely balanced, complex, multi-step process.

Some of stem cells are now in clinical use, such as embryonic stem cells (ESCs) derived from the inner cell mass of preimplantation embryos and induced pluripotent stem cells (iPSCs) derived from the epiblast layer of implanted embryos [45, 46]. Mesenchymal stem cells (MSCs) can promote tissue repair through the secretion of extracellular vesicles that carry a variety of cytokines, growth factors and microRNAs (miRNAs) [47]. Adipose tissue-derived stem cells (ADSCs) are a replacement therapy for MSCs, with the similar mechanism which secretion extracellular vesicles (EVs) to multiple proteins possessing neuroprotective and neurogenesis activities [48]. NSCs participate extensively in mammalian brain homeostasis and repair and exhibit pleiotropic intrinsic properties which makes them a good method for the treatment of AD [49].

3.1 ESCs

ESCs are cells isolated from early embryos or primitive gonads. It has the characteristics of infinite proliferation, self-renewal and multidirectional differentiation in vitro culture. Both in vitro and in vivo, ESCs can be induced to differentiate into almost all cell types in the body, so they can be used to improve the recovery of neurodegenerative diseases (such as AD). Therefore ESCs have a broad application prospect in autologous stem cell therapy [50, 51]. Thymic epithelial progenitor cells derived from mouse ESCs with deleted amyloid precursor protein gene have been proved to have the ability to alleviate AD symptoms [52]. Early human embryonic stem cells (hESCs)-derived neural populations consist of various embryonic neural progenitors (ENPs) with broad neural developmental propensity. The hESC-ENP-enriched neural transcription factors (TFs) can directly transform human cells into ENP phenotypes. Induced ENPs (IENPs) and their derivatives summarize the signature pathological characteristics of AD and hold promise for future strategies for disease modeling and clinical intervention [53].

Although ESCs are good candidates for AD cell therapy, they may bring some ethical and practical problems. Even if we overcome the problem of immune rejection, there have been reports of teratomas resulting from transplanted ESCs [54].

3.2 NSCs

NSCs have the ability to differentiate into neuronal astrocytes and oligodendrocytes, which are self-renewing and sufficient to provide a large number of brain tissue cells [55, 56]. In the past, it was thought that NSCs lost their ability to regenerate during the prenatal period or several months after birth. However, some recent studies have shown that NSCs also exist in adult brain tissues, mainly located in the subventricular zone (SVZ) and hippocampus dentate gyrus (DG) [57, 58, 59, 60]. Due to their multidirectional differentiation and self-renewal, NSCs play an important role in maintaining brain homeostasis, promoting normal nerve development and repairing damaged nerves, which provides a possible choice for stem cell therapy for AD [49, 61]. A large number of studies have shown that the gradual accumulation of Aβ leading to the loss of synapses related to cognitive deficits is an important mechanism of AD [62]. In the hippocampus of AD mice after NSCs transplantation, the level of Synaptophysin (SYP), postsynaptic density protein 95 (PSD-95) and microtubule-associated protein (MAP-2) were significantly increased, which are important protein markers related to synaptic plasticity and play an important role in synaptic plasticity and stability, indicating improved learning and memory ability in AD mice [63, 64, 65]. Damage of cholinergic neurons in the basal forebrain is another important feature of AD [66]. Reduced cholinergic function due to cholinergic neuron injury may results in learning and memory impairments [67]. Transplantation of NSCs into the basal forebrain will increase the level of choline acetyltransferase (ChAT) protein, restoring the damaged neurons and improving the learning and memory ability [68, 69].

Recent studies have demonstrated the mechanism of NSCs transplantation to improve cognitive function, which is replacing damaged neurons with the differentiation of transplanted NSCs and enhancing synaptic density by releasing neurotrophic factors [61, 70, 71]. Neurotrophic factors have been shown to improve cognitive impairment [72, 73]. Although NSCs transplantation has great potential to be an excellent choice of cell therapy for AD in the future, there are many problems in its application: (1) The attribution that supports the differentiation of NSCs into a specific cell type is not clear. (2) Although NSCs transplantation can salvage synaptic damage and participate in the interaction of endogenous neuronal circuit function, there is no accurate answer to the duration of this effect. (3) The localization of the transplanted area and the viability of the transplanted cells are only the initial challenges of NSCs therapy, and subsequent interactions with cells in the host environment are also important. In some studies, NSCs after transplantation is difficult to trace, and in the cases where NSCs can be traced, the number of activated cells is also difficult to quantify [74]. (4) Many studies have identified transplanted NSCs have potential risk of developing brain tumors, such as glioblastoma [75, 76]. (5) Extrinsic NSCs transplantation also involves ethical issues. Direct isolation of NSCs from the primary tissue is dangerous. Non-patient-specific NSCs are more likely to result in immune rejection [54, 77].

3.3 MSCs

MSCs are pluripotent stem cells, which have all the common features of stem cells, namely self-renewal and multidirectional differentiation. As major stem cells that have undergone extensive clinical trials, MSCs bring hope for the treatment of a variety of diseases [78]. MSCs come from a wide range of sources. The most common ones are bone marrow mesenchymal stem cells (BMSCs), adipose-derived stem cells (ADSCs), umbilical cord derived mesenchymal stem cells (UC-MSCs), etc. Their biological characteristics are also different [79].

MSCs have the ability of immune regulation, neuroprotection and regeneration. The main mechanisms of MSCs in the treatment of AD are as follows [80]: (1) Secrete growth factors: MSCs secrete a variety of pro-cytokines that may play a beneficial role in AD [81]. (2) Secrete exosomes: Exosomes refer to extracellular vesicles, which are biocompatible nanoparticles with lipid membranes. These vesicles can transmit messages across biological barriers. Studies have shown that intercellular exchange of miRNA and proteins through EVs can reduce neuroinflammation, promote neurogenesis and angiogenesis, save learning disabilities and improve functional recovery [82, 83]. (3) Reduce neuroinflammation by regulating autophagy: MSCs can affect the autophagy of immune cells involved in injury-induced inflammation, thereby reducing their survival, proliferation and function, and facilitating the regression of inflammation. In addition, MSCs can affect the autophagy of endogenous adult or progenitor cells, promote their survival, proliferation and differentiation, and support the recovery of functional tissues [84]. In addition, foreign proteins conveyed by MSCs can regulate microglia function and enhance neurogenesis, so as to alleviate early memory deficits in AD [85]. Transplantation of MSCs carrying CX3CL1 (a multifunctional inflammatory chemokine with a single receptor CX3CR1) [86] and Wnt3a (CX3CL1-Wnt3a-MSC) can regulate phosphoinositide 3-kinase/activated protein kinase B (PI3K/AKT) signaling to inhibit the activity of glycogen synthase kinase 3 beta (GSK3β), improving the neurobehavioral function of mice by transplanting microglia with neurotoxicity and promoting hippocampal neurogenesis.

Reports have shown that EVs secreted by adipocytes derived from ADSCs may treat AD by alleviating neuronal damage, promoting neurogenesis and reducing the increase of neuronal apoptosis [48, 87]. EVs secreted by BMSCs can reach astrocytes to promote synaptic development and improve cognitive impairment [88, 89]. Hepatocyte growth factor (HGF), a core functional factor secreted by UC-MSCs, plays a key role in regulating the recovery of damaged nerve cells [90]. MSCs derived from ESCs have a better effect than BMSCs in the treatment of AD [91].

Modified MSCs pretreated with different conditions or reagents can significantly enhance the therapeutic effect of AD and improve cognitive impairment, such as cytokine pretreated MSCs [92], hypoxia preconditioned MSCs (PCMSCs) [93], MSCs modified by lin28B [94], MSCs prepared by cerebrospinal fluid of AD patients [95], ADSCs pretreated by melatonin (MT) [96], UC-MSCs combined with resveratrol [97].

3.4 iPSCs

Using defined reprogramming factors to reprogram fully differentiated somatic cells into iPSCs has become a novel strategy to produce pluripotent cells derived from patients that enable autologous transplantation [98]. The apolipoprotein E4 (ApoE4) variant is the single greatest genetic risk factor for sporadic Alzheimer’s disease (sAD) [27, 28, 29, 30]. sAD iPSCs convert ApoE4 to ApoE3 in brain cell types. This conversion can reduce many AD-related diseases [99]. The generation of neural precursors from iPSCs has also been extensively studied. In the production of astrocytes, the mutation in presenilin1 (PSEN1) increased Aβ production and oxidative stress. At the same time, it also altered cytokine release and Ca2+ homeostasis. These changes reducing neuronal support function in PSEN1 astrocytes [100, 101]. EVs of either 50–200 nm in size (called exosomes) or 200 nm−1 μm in size (called micro-vesicles) are membrane-bounded vesicles. They can carry RNAs, proteins, and other metabolites. They are secreted from all cell types and present in biological fluids such as serum and plasma [50, 102]. Human iPSCs can be cultured infinitely under a chemically defined medium. The properties and functions of exosomes and micro-vesicles (called EMVs) from human iPSCs are different with the ones secreted by human MSCs. Purified EVs produced by both stem cell types have similar sizes, but human iPSCs produced 16-fold more EVs than MSCs [103]. Neurons from patients with early-onset familial Alzheimer’s disease (fAD) and patients with late-onset sAD showed increased phosphorylation of Tau protein at all investigated phosphorylation sites. Relative to the control neurons, neurons derived from patients with fAD and patients with sAD exhibited higher levels of extracellular amyloid-β 1–40 (Aβ1–40) and amyloid-β 1–42 (Aβ1–42) [104, 105, 106]. Using iPSCs-derived neurons to recapitulate AD pathology in vitro has significant applications in the study of pathogenesis and screening for potential therapeutic drugs. They are now the subject of extensive study in vitro [107]. Studies have also shown that EVs from iPSCs can play an important role in heart repair [108].

3.5 Clinical trials and results in humans

Due to the inconsistent results of various preclinical studies, stem cell therapies other than MSCs are still difficult to be applied clinically. Some articles specifically showed us the application of MSCs-based stem cell therapy in human clinical trials [6, 80, 109, 110]. In recent years, more studies have been conducted on rodents. The effects of MSCs on AD pathology and cognitive mouse models may be mediated by the regulation of neuroinflammation [111, 112]. In recent years, clinical trials using mesenchymal stem cells have been conducted around the world. A completed clinical trial in the United States (Trial identifier: NCT03117738) investigated the safety and efficacy of autologous ADSCs. At the same time, a team studied the efficacy of UC-MSCs (Trial identifier: NCT01297218). Compared with cholinergic drugs that only improve symptoms, UC-MSCs are immunologically stable and not-toxic, and have better therapeutic effect on AD. UC-MSCS remain a common cell choice, although there are key differences in cell number, dose quantity, and dose schedule (Trial identifier: NCT03172117). Two separate trials, both currently undergoing recruitment, will utilize alternative MSC sources. One studies human MSCs (Trial identifier: NCT02833792) and evaluates its safety and efficacy. The other utilizes the exosomes derived from allogenic adipose mesenchymal stem cells (MSCs-Exos) (Trial identifier: NCT04388982) to treat patients with mild to moderate dementia due to AD. While many of these trials employ an intravenous infusion administration route, one trial (Trial identifier: NCT03724136) administered BMSCs to the nasal mucosa topically, to investigate whether there was an improvement in efficacy in combination with intravenous injection.

Advertisement

4. Future directions

Numerous preclinical studies have revealed the different mechanisms of various stem cells and demonstrated the great potential of stem cells to treat AD. However, the biggest problem in this area of research is that it is difficult to translate animal studies into human trials. In fact, researchers have used nearly a hundred methods to effectively treat AD in transgenic mouse models. Disappointingly, almost every approach has failed in human clinical trials or has never even been tested in humans. Clearly, rodent models and their pathological assumptions are insufficient to predict clinical outcomes in humans. Therefore, the establishment of more accurate models is needed for cell therapy of AD. Since the goal of truly simulating the pathological progress of AD in human body has been achieved, more experiments on cell therapy need to be carried out.

At the same time, key questions remain to be addressed, including the safety of treatment, optimal cell source and delivery system. While cell therapies may not be able to fully compensate for the loss of extensive synapses, they can help to temporarily improve existing depleted circuits enough to improve cognitive function, restore basic daily living functions, and improve quality of life. For us, stem cell therapy for AD still has a long way to go.

Advertisement

5. Conclusion

AD is a neurodegenerative disease, which is characterized by excessive deposition of Aβ and abnormal phosphorylation of Tau protein and synaptic loss. Studies and clinical trials in recent years are also based on these basic mechanisms. Although the role of stem cell therapy in AD is not fully understood, many preclinical studies have provided a number of promising results. However, human clinical trials are still in their infancy, and most current research is still centered on animal experiments. But it also shows the broad prospects of stem cell therapy for the AD. A large number of preclinical studies have demonstrated the theoretical basis, and new studies are continuing to reveal the underlying mechanisms. Among many stem cells, MSCs-based therapies are widely accepted and have met certain clinical trial standards. The vast majority of cell therapies for AD have been conducted on rodents, and we must be aware of a wide range of physiological differences between humans and rodents. We need to understand the mechanism of treatment through animal experiments and establish the correct translation model for human application.

References

  1. 1. Vasic, V.; Barth, K.; Schmidt, M. H. H. Neurodegeneration and Neuro-Regeneration-Alzheimer's Disease and Stem Cell Therapy. Int J Mol Sci 2019, 20
  2. 2. Zhang, H.; Zheng, Y. [beta Amyloid Hypothesis in Alzheimer's Disease: Pathogenesis, Prevention, and Management]. Zhongguo Yi Xue Ke Xue Yuan Xue Bao 2019, 41, 702-708
  3. 3. Soria Lopez, J. A.; Gonzalez, H. M.; Leger, G. C. Alzheimer's disease. Handb Clin Neurol 2019, 167, 231-255
  4. 4. Esquerda-Canals, G.; Montoliu-Gaya, L.; Guell-Bosch, J.; Villegas, S. Mouse Models of Alzheimer's Disease. J Alzheimers Dis 2017, 57, 1171-1183
  5. 5. Breijyeh, Z.; Karaman, R. Comprehensive Review on Alzheimer's Disease: Causes and Treatment. Molecules 2020, 25
  6. 6. Duncan, T.; Valenzuela, M. Alzheimer's disease, dementia, and stem cell therapy. Stem Cell Res Ther 2017, 8, 111
  7. 7. Reiss, A. B.; Arain, H. A.; Stecker, M. M.; Siegart, N. M.; Kasselman, L. J. Amyloid toxicity in Alzheimer's disease. Rev Neurosci 2018, 29, 613-627
  8. 8. Tiwari, S.; Atluri, V.; Kaushik, A.; Yndart, A.; Nair, M. Alzheimer's disease: pathogenesis, diagnostics, and therapeutics. Int J Nanomedicine 2019, 14, 5541-5554
  9. 9. Gallardo, G.; Holtzman, D. M. Amyloid-beta and Tau at the Crossroads of Alzheimer's Disease. Adv Exp Med Biol 2019, 1184, 187-203
  10. 10. Guo, T.; Zhang, D.; Zeng, Y.; Huang, T. Y.; Xu, H.; Zhao, Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer's disease. Mol Neurodegener 2020, 15, 40
  11. 11. Shimada, H.; Kitamura, S.; Shinotoh, H.; Endo, H.; Niwa, F.; Hirano, S.; Kimura, Y.; Zhang, M. R.; Kuwabara, S.; Suhara, T.; Higuchi, M. Association between Abeta and tau accumulations and their influence on clinical features in aging and Alzheimer's disease spectrum brains: A [(11)C]PBB3-PET study. Alzheimers Dement (Amst) 2017, 6, 11-20
  12. 12. Chen, Y. G. Research Progress in the Pathogenesis of Alzheimer's Disease. Chin Med J (Engl) 2018, 131, 1618-1624
  13. 13. Lauretti, E.; Li, J. G.; Di Meco, A.; Pratico, D. Glucose deficit triggers tau pathology and synaptic dysfunction in a tauopathy mouse model. Transl Psychiatry 2017, 7, e1020
  14. 14. Lee, S. H.; Le Pichon, C. E.; Adolfsson, O.; Gafner, V.; Pihlgren, M.; Lin, H.; Solanoy, H.; Brendza, R.; Ngu, H.; Foreman, O.; Chan, R.; Ernst, J. A.; DiCara, D.; Hotzel, I.; Srinivasan, K.; Hansen, D. V.; Atwal, J.; Lu, Y.; Bumbaca, D.; Pfeifer, A.; Watts, R. J.; Muhs, A.; Scearce-Levie, K.; Ayalon, G. Antibody-Mediated Targeting of Tau In Vivo Does Not Require Effector Function and Microglial Engagement. Cell Rep 2016, 16, 1690-1700
  15. 15. Ittner, A.; Chua, S. W.; Bertz, J.; Volkerling, A.; van der Hoven, J.; Gladbach, A.; Przybyla, M.; Bi, M.; van Hummel, A.; Stevens, C. H.; Ippati, S.; Suh, L. S.; Macmillan, A.; Sutherland, G.; Kril, J. J.; Silva, A. P.; Mackay, J. P.; Poljak, A.; Delerue, F.; Ke, Y. D.; Ittner, L. M. Site-specific phosphorylation of tau inhibits amyloid-beta toxicity in Alzheimer's mice. Science 2016, 354, 904-908
  16. 16. Jeong, S. Molecular and Cellular Basis of Neurodegeneration in Alzheimer's Disease. Mol Cells 2017, 40, 613-620
  17. 17. Uddin, M. S.; Stachowiak, A.; Mamun, A. A.; Tzvetkov, N. T.; Takeda, S.; Atanasov, A. G.; Bergantin, L. B.; Abdel-Daim, M. M.; Stankiewicz, A. M. Autophagy and Alzheimer's Disease: From Molecular Mechanisms to Therapeutic Implications. Front Aging Neurosci 2018, 10, 04
  18. 18. Skaper, S. D.; Facci, L.; Zusso, M.; Giusti, P. Synaptic Plasticity, Dementia and Alzheimer Disease. CNS Neurol Disord Drug Targets 2017, 16, 220-233
  19. 19. Colom-Cadena, M.; Spires-Jones, T.; Zetterberg, H.; Blennow, K.; Caggiano, A.; DeKosky, S. T.; Fillit, H.; Harrison, J. E.; Schneider, L. S.; Scheltens, P.; de Haan, W.; Grundman, M.; van Dyck, C. H.; Izzo, N. J.; Catalano, S. M.; Synaptic Health Endpoints Working, G. The clinical promise of biomarkers of synapse damage or loss in Alzheimer's disease. Alzheimers Res Ther2020, 12, 21
  20. 20. Cai, Q.; Tammineni, P. Mitochondrial Aspects of Synaptic Dysfunction in Alzheimer's Disease. J Alzheimers Dis 2017, 57, 1087-1103
  21. 21. Talantova, M.; Sanz-Blasco, S.; Zhang, X.; Xia, P.; Akhtar, M. W.; Okamoto, S.; Dziewczapolski, G.; Nakamura, T.; Cao, G.; Pratt, A. E.; Kang, Y. J.; Tu, S.; Molokanova, E.; McKercher, S. R.; Hires, S. A.; Sason, H.; Stouffer, D. G.; Buczynski, M. W.; Solomon, J. P.; Michael, S.; Powers, E. T.; Kelly, J. W.; Roberts, A.; Tong, G.; Fang-Newmeyer, T.; Parker, J.; Holland, E. A.; Zhang, D.; Nakanishi, N.; Chen, H. S.; Wolosker, H.; Wang, Y.; Parsons, L. H.; Ambasudhan, R.; Masliah, E.; Heinemann, S. F.; Pina-Crespo, J. C.; Lipton, S. A. Abeta induces astrocytic glutamate release, extrasynaptic NMDA receptor activation, and synaptic loss. Proc Natl Acad Sci U S A2013, 110, E2518-2527
  22. 22. Park, J. C.; Han, S. H.; Mook-Jung, I. Peripheral inflammatory biomarkers in Alzheimer's disease: a brief review. BMB Rep 2020, 53, 10-19
  23. 23. Regen, F.; Hellmann-Regen, J.; Costantini, E.; Reale, M. Neuroinflammation and Alzheimer's Disease: Implications for Microglial Activation. Curr Alzheimer Res 2017, 14, 1140-1148
  24. 24. Kloske, C. M.; Wilcock, D. M. The Important Interface Between Apolipoprotein E and Neuroinflammation in Alzheimer's Disease. Front Immunol 2020, 11, 754
  25. 25. Houtman, J.; Freitag, K.; Gimber, N.; Schmoranzer, J.; Heppner, F. L.; Jendrach, M. Beclin1-driven autophagy modulates the inflammatory response of microglia via NLRP3. EMBO J 2019, 38
  26. 26. Liddelow, S. A.; Guttenplan, K. A.; Clarke, L. E.; Bennett, F. C.; Bohlen, C. J.; Schirmer, L.; Bennett, M. L.; Munch, A. E.; Chung, W. S.; Peterson, T. C.; Wilton, D. K.; Frouin, A.; Napier, B. A.; Panicker, N.; Kumar, M.; Buckwalter, M. S.; Rowitch, D. H.; Dawson, V. L.; Dawson, T. M.; Stevens, B.; Barres, B. A. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481-487
  27. 27. Yin, Y.; Wang, Z. ApoE and Neurodegenerative Diseases in Aging. Adv Exp Med Biol 2018, 1086, 77-92
  28. 28. Yamazaki, Y.; Zhao, N.; Caulfield, T. R.; Liu, C. C.; Bu, G. Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol 2019, 15, 501-518
  29. 29. Serrano-Pozo, A.; Das, S.; Hyman, B. T. APOE and Alzheimer's disease: advances in genetics, pathophysiology, and therapeutic approaches. Lancet Neurol 2021, 20, 68-80
  30. 30. Zhao, N.; Liu, C. C.; Qiao, W.; Bu, G. Apolipoprotein E, Receptors, and Modulation of Alzheimer's Disease. Biol Psychiatry 2018, 83, 347-357
  31. 31. Huat, T. J.; Camats-Perna, J.; Newcombe, E. A.; Valmas, N.; Kitazawa, M.; Medeiros, R. Metal Toxicity Links to Alzheimer's Disease and Neuroinflammation. J Mol Biol 2019, 431, 1843-1868
  32. 32. Ahmed, T.; Zahid, S.; Mahboob, A.; Farhat, S. M. Cholinergic System and Post-translational Modifications: An Insight on the Role in Alzheimer's Disease. Curr Neuropharmacol 2017, 15, 480-494
  33. 33. Haam, J.; Yakel, J. L. Cholinergic modulation of the hippocampal region and memory function. J Neurochem2017, 142 Suppl 2, 111-121
  34. 34. Pepeu, G.; Grazia Giovannini, M. The fate of the brain cholinergic neurons in neurodegenerative diseases. Brain Res 2017, 1670, 173-184
  35. 35. Du, X.; Wang, X.; Geng, M. Alzheimer's disease hypothesis and related therapies. Transl Neurodegener 2018, 7, 2
  36. 36. 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
  37. 37. Pramanik, S.; Sulistio, Y. A.; Heese, K. Neurotrophin Signaling and Stem Cells-Implications for Neurodegenerative Diseases and Stem Cell Therapy. Mol Neurobiol 2017, 54, 7401-7459
  38. 38. Skop, N. B.; Singh, S.; Antikainen, H.; Saqcena, C.; Calderon, F.; Rothbard, D. E.; Cho, C. H.; Gandhi, C. D.; Levison, S. W.; Dobrowolski, R. Subacute Transplantation of Native and Genetically Engineered Neural Progenitors Seeded on Microsphere Scaffolds Promote Repair and Functional Recovery After Traumatic Brain Injury. ASN Neuro 2019, 11, 1759091419830186
  39. 39. Karimipour, M.; Rahbarghazi, R.; Tayefi, H.; Shimia, M.; Ghanadian, M.; Mahmoudi, J.; Bagheri, H. S. Quercetin promotes learning and memory performance concomitantly with neural stem/progenitor cell proliferation and neurogenesis in the adult rat dentate gyrus. Int J Dev Neurosci 2019, 74, 18-26
  40. 40. Wen, C.; Huang, C.; Yang, M.; Fan, C.; Li, Q.; Zhao, J.; Gan, D.; Li, A.; Zhu, L.; Lu, D. The Secretion from Bone Marrow Mesenchymal Stem Cells Pretreated with Berberine Rescues Neurons with Oxidative Damage Through Activation of the Keap1-Nrf2-HO-1 Signaling Pathway. Neurotox Res 2020, 38, 59-73
  41. 41. Vymetalova, L.; Kucirkova, T.; Knopfova, L.; Pospisilova, V.; Kasko, T.; Lejdarova, H.; Makaturova, E.; Kuglik, P.; Oralova, V.; Matalova, E.; Benes, P.; Koristek, Z.; Forostyak, S. Large-Scale Automated Hollow-Fiber Bioreactor Expansion of Umbilical Cord-Derived Human Mesenchymal Stromal Cells for Neurological Disorders. Neurochem Res 2020, 45, 204-214
  42. 42. Morelli, A.; Sarchielli, E.; Guarnieri, G.; Coppi, E.; Pantano, D.; Comeglio, P.; Nardiello, P.; Pugliese, A. M.; Ballerini, L.; Matucci, R.; Ambrosini, S.; Castronovo, G.; Valente, R.; Mazzanti, B.; Bucciantini, S.; Maggi, M.; Casamenti, F.; Gallina, P.; Vannelli, G. B. Young Human Cholinergic Neurons Respond to Physiological Regulators and Improve Cognitive Symptoms in an Animal Model of Alzheimer's Disease. Front Cell Neurosci 2017, 11, 339
  43. 43. Jin, K.; Zhu, Y.; Sun, Y.; Mao, X. O.; Xie, L.; Greenberg, D. A. Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo. Proc Natl Acad Sci U S A 2002, 99, 11946-11950
  44. 44. Li, B.; Gao, Y.; Zhang, W.; Xu, J. R. Regulation and effects of neurotrophic factors after neural stem cell transplantation in a transgenic mouse model of Alzheimer disease. J Neurosci Res 2018, 96, 828-840
  45. 45. Bloor, A. J. C.; Patel, A.; Griffin, J. E.; Gilleece, M. H.; Radia, R.; Yeung, D. T.; Drier, D.; Larson, L. S.; Uenishi, G. I.; Hei, D.; Kelly, K.; Slukvin, I.; Rasko, J. E. J. Production, safety and efficacy of iPSC-derived mesenchymal stromal cells in acute steroid-resistant graft versus host disease: a phase I, multicenter, open-label, dose-escalation study. Nat Med 2020, 26, 1720-1725
  46. 46. Morishima, Y.; Morishima, S.; Murata, M.; Arima, N.; Uchida, N.; Sugio, Y.; Takahashi, S.; Matsuhashi, Y.; Onizuka, M.; Eto, T.; Nagafuji, K.; Onishi, Y.; Inoue, M.; Atsuta, Y.; Fukuda, T.; Ichinohe, T.; Kato, S.; Kanda, J. Impact of Homozygous Conserved Extended HLA Haplotype on Single Cord Blood Transplantation: Lessons for Induced Pluripotent Stem Cell Banking and Transplantation in Allogeneic Settings. Biol Blood Marrow Transplant 2020, 26, 132-138
  47. 47. Keshtkar, S.; Azarpira, N.; Ghahremani, M. H. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther 2018, 9, 63
  48. 48. Ma, X.; Huang, M.; Zheng, M.; Dai, C.; Song, Q.; Zhang, Q.; Li, Q.; Gu, X.; Chen, H.; Jiang, G.; Yu, Y.; Liu, X.; Li, S.; Wang, G.; Chen, H.; Lu, L.; Gao, X. ADSCs-derived extracellular vesicles alleviate neuronal damage, promote neurogenesis and rescue memory loss in mice with Alzheimer's disease. J Control Release 2020, 327, 688-702
  49. 49. 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
  50. 50. 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
  51. 51. De Filippis, L.; Zalfa, C.; Ferrari, D. Neural Stem Cells and Human Induced Pluripotent Stem Cells to Model Rare CNS Diseases. CNS Neurol Disord Drug Targets 2017, 16, 915-926
  52. 52. 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
  53. 53. Hou, P. S.; Chuang, C. Y.; Yeh, C. H.; Chiang, W.; Liu, H. J.; Lin, T. N.; Kuo, H. C. Direct Conversion of Human Fibroblasts into Neural Progenitors Using Transcription Factors Enriched in Human ESC-Derived Neural Progenitors. Stem Cell Reports 2017, 8, 54-68
  54. 54. Sugaya, K.; Vaidya, M. Stem Cell Therapies for Neurodegenerative Diseases. Adv Exp Med Biol 2018, 1056, 61-84
  55. 55. Han, F.; Bi, J.; Qiao, L.; Arancio, O. Stem Cell Therapy for Alzheimer's Disease. Adv Exp Med Biol 2020, 1266, 39-55
  56. 56. Wong, R. S. Y.; Cheong, S. K. Therapeutic potentials of neural stem cells in Alzheimer's disease. Malays J Pathol 2020, 42, 157-170
  57. 57. Zhang, W.; Gu, G. J.; Zhang, Q.; Liu, J. H.; Zhang, B.; Guo, Y.; Wang, M. Y.; Gong, Q. Y.; Xu, J. R. NSCs promote hippocampal neurogenesis, metabolic changes and synaptogenesis in APP/PS1 transgenic mice. Hippocampus 2017, 27, 1250-1263
  58. 58. Bond, A. M.; Ming, G. L.; Song, H. Adult Mammalian Neural Stem Cells and Neurogenesis: Five Decades Later. Cell Stem Cell 2015, 17, 385-395
  59. 59. Guo, W.; Patzlaff, N. E.; Jobe, E. M.; Zhao, X. Isolation of multipotent neural stem or progenitor cells from both the dentate gyrus and subventricular zone of a single adult mouse. Nat Protoc 2012, 7, 2005-2012
  60. 60. Lee, J. P.; McKercher, S.; Muller, F. J.; Snyder, E. Y. Neural stem cell transplantation in mouse brain. Curr Protoc Neurosci2008, Chapter 3, Unit 3 10
  61. 61. Marsh, S. E.; Blurton-Jones, M. Neural stem cell therapy for neurodegenerative disorders: The role of neurotrophic support. Neurochem Int 2017, 106, 94-100
  62. 62. Shankar, G. M.; Li, S.; Mehta, T. H.; Garcia-Munoz, A.; Shepardson, N. E.; Smith, I.; Brett, F. M.; Farrell, M. A.; Rowan, M. J.; Lemere, C. A.; Regan, C. M.; Walsh, D. M.; Sabatini, B. L.; Selkoe, D. J. Amyloid-beta protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory. Nat Med 2008, 14, 837-842
  63. 63. 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
  64. 64. Schmitt, U.; Tanimoto, N.; Seeliger, M.; Schaeffel, F.; Leube, R. E. Detection of behavioral alterations and learning deficits in mice lacking synaptophysin. Neuroscience 2009, 162, 234-243
  65. 65. Tu, S.; Okamoto, S.; Lipton, S. A.; Xu, H. Oligomeric Abeta-induced synaptic dysfunction in Alzheimer's disease. Mol Neurodegener 2014, 9, 48
  66. 66. Hampel, H.; Mesulam, M. M.; Cuello, A. C.; Farlow, M. R.; Giacobini, E.; Grossberg, G. T.; Khachaturian, A. S.; Vergallo, A.; Cavedo, E.; Snyder, P. J.; Khachaturian, Z. S. The cholinergic system in the pathophysiology and treatment of Alzheimer's disease. Brain 2018, 141, 1917-1933
  67. 67. Solari, N.; Hangya, B. Cholinergic modulation of spatial learning, memory and navigation. Eur J Neurosci 2018, 48, 2199-2230
  68. 68. 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, Abeta Elimination, and Neuroregeneration in APPswe/PS1dE9 Mice. Int J Mol Sci 2020, 21
  69. 69. Chen, Y.; Pan, C.; Xuan, A.; Xu, L.; Bao, G.; Liu, F.; Fang, J.; Long, D. Treatment Efficacy of NGF Nanoparticles Combining Neural Stem Cell Transplantation on Alzheimer's Disease Model Rats. Med Sci Monit 2015, 21, 3608-3615
  70. 70. Wu, C. C.; Lien, C. C.; Hou, W. H.; Chiang, P. M.; Tsai, K. J. Gain of BDNF Function in Engrafted Neural Stem Cells Promotes the Therapeutic Potential for Alzheimer's Disease. Sci Rep 2016, 6, 27358
  71. 71. Kim, D. H.; Lim, H.; Lee, D.; Choi, S. J.; Oh, W.; Yang, Y. S.; Oh, J. S.; Hwang, H. H.; Jeon, H. B. Thrombospondin-1 secreted by human umbilical cord blood-derived mesenchymal stem cells rescues neurons from synaptic dysfunction in Alzheimer's disease model. Sci Rep 2018, 8, 354
  72. 72. Di Carlo, P.; Punzi, G.; Ursini, G. Brain-derived neurotrophic factor and schizophrenia. Psychiatr Genet 2019, 29, 200-210
  73. 73. Lima Giacobbo, B.; Doorduin, J.; Klein, H. C.; Dierckx, R.; Bromberg, E.; de Vries, E. F. J. Brain-Derived Neurotrophic Factor in Brain Disorders: Focus on Neuroinflammation. Mol Neurobiol 2019, 56, 3295-3312
  74. 74. Zheng, Y.; Huang, J.; Zhu, T.; Li, R.; Wang, Z.; Ma, F.; Zhu, J. Stem Cell Tracking Technologies for Neurological Regenerative Medicine Purposes. Stem Cells Int 2017, 2017, 2934149
  75. 75. Gimple, R. C.; Bhargava, S.; Dixit, D.; Rich, J. N. Glioblastoma stem cells: lessons from the tumor hierarchy in a lethal cancer. Genes Dev 2019, 33, 591-609
  76. 76. Yelle, N.; Bakhshinyan, D.; Venugopal, C.; Singh, S. K. Introduction to Brain Tumor Stem Cells. Methods Mol Biol 2019, 1869, 1-9
  77. 77. 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
  78. 78. Wei, X.; Yang, X.; Han, Z. P.; Qu, F. F.; Shao, L.; Shi, Y. F. Mesenchymal stem cells: a new trend for cell therapy. Acta Pharmacol Sin 2013, 34, 747-754
  79. 79. Hass, R.; Kasper, C.; Bohm, S.; Jacobs, R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal 2011, 9, 12
  80. 80. Staff, N. P.; Jones, D. T.; Singer, W. Mesenchymal Stromal Cell Therapies for Neurodegenerative Diseases. Mayo Clin Proc 2019, 94, 892-905
  81. 81. Park, S. E.; Lee, N. K.; Na, D. L.; Chang, J. W. Optimal mesenchymal stem cell delivery routes to enhance neurogenesis for the treatment of Alzheimer's disease: optimal MSCs delivery routes for the treatment of AD. Histol Histopathol 2018, 33, 533-541
  82. 82. Reza-Zaldivar, E. E.; Hernandez-Sapiens, M. A.; Minjarez, B.; Gutierrez-Mercado, Y. K.; Marquez-Aguirre, A. L.; Canales-Aguirre, A. A. Potential Effects of MSC-Derived Exosomes in Neuroplasticity in Alzheimer's Disease. Front Cell Neurosci 2018, 12, 317
  83. 83. Izadpanah, M.; Dargahi, L.; Ai, J.; Asgari Taei, A.; Ebrahimi Barough, S.; Mowla, S. J.; TavoosiDana, G.; Farahmandfar, M. Extracellular Vesicles as a Neprilysin Delivery System Memory Improvement in Alzheimer's Disease. Iran J Pharm Res 2020, 19, 45-60
  84. 84. Ceccariglia, S.; Cargnoni, A.; Silini, A. R.; Parolini, O. Autophagy: a potential key contributor to the therapeutic action of mesenchymal stem cells. Autophagy 2020, 16, 28-37
  85. 85. Li, A.; Zhao, J.; Fan, C.; Zhu, L.; Huang, C.; Li, Q.; Gan, D.; Wen, C.; Chen, M.; Lu, D. Delivery of exogenous proteins by mesenchymal stem cells attenuates early memory deficits in a murine model of Alzheimer's disease. Neurobiol Aging 2020, 86, 81-91
  86. 86. Conroy, M. J.; Lysaght, J. CX3CL1 Signaling in the Tumor Microenvironment. Adv Exp Med Biol 2020, 1231, 1-12
  87. 87. Lee, M.; Ban, J. J.; Yang, S.; Im, W.; Kim, M. The exosome of adipose-derived stem cells reduces beta-amyloid pathology and apoptosis of neuronal cells derived from the transgenic mouse model of Alzheimer's disease. Brain Res 2018, 1691, 87-93
  88. 88. 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
  89. 89. Elia, C. A.; Tamborini, M.; Rasile, M.; Desiato, G.; Marchetti, S.; Swuec, P.; Mazzitelli, S.; Clemente, F.; Anselmo, A.; Matteoli, M.; Malosio, M. L.; Coco, S. Intracerebral Injection of Extracellular Vesicles from Mesenchymal Stem Cells Exerts Reduced Abeta Plaque Burden in Early Stages of a Preclinical Model of Alzheimer's Disease. Cells 2019, 8
  90. 90. Jia, Y.; Cao, N.; Zhai, J.; Zeng, Q.; Zheng, P.; Su, R.; Liao, T.; Liu, J.; Pei, H.; Fan, Z.; Zhou, J.; Xi, J.; He, L.; Chen, L.; Nan, X.; Yue, W.; Pei, X. HGF Mediates Clinical-Grade Human Umbilical Cord-Derived Mesenchymal Stem Cells Improved Functional Recovery in a Senescence-Accelerated Mouse Model of Alzheimer's Disease. Adv Sci (Weinh) 2020, 7, 1903809
  91. 91. 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 Alzheimers Dis 2020, 76, 1281-1296
  92. 92. Losurdo, M.; Pedrazzoli, M.; D'Agostino, C.; Elia, C. A.; Massenzio, F.; Lonati, E.; Mauri, M.; Rizzi, L.; Molteni, L.; Bresciani, E.; Dander, E.; D'Amico, G.; Bulbarelli, A.; Torsello, A.; Matteoli, M.; Buffelli, M.; Coco, S. Intranasal delivery of mesenchymal stem cell-derived extracellular vesicles exerts immunomodulatory and neuroprotective effects in a 3xTg model of Alzheimer's disease. Stem Cells Transl Med 2020, 9, 1068-1084
  93. 93. Cui, G. H.; Wu, J.; Mou, F. F.; Xie, W. H.; Wang, F. B.; Wang, Q. L.; Fang, J.; Xu, Y. W.; Dong, Y. R.; Liu, J. R.; Guo, H. D. Exosomes derived from hypoxia-preconditioned mesenchymal stromal cells ameliorate cognitive decline by rescuing synaptic dysfunction and regulating inflammatory responses in APP/PS1 mice. FASEB J 2018, 32, 654-668
  94. 94. Wu, K.; Zhang, R.; Lu, Y.; Wen, L.; Li, Y.; Duan, R.; Yao, Y.; Jia, Y. Lin28B regulates the fate of grafted mesenchymal stem cells and enhances their protective effects against Alzheimer's disease by upregulating IGF-2. J Cell Physiol 2019, 234, 21860-21876
  95. 95. Lee, J.; Kwon, S. J.; Kim, J. H.; Jang, H.; Lee, N. K.; Hwang, J. W.; Kim, J. H.; Chang, J. W.; Na, D. L. Cerebrospinal fluid from Alzheimer's disease patients as an optimal formulation for therapeutic application of mesenchymal stem cells in Alzheimer's disease. Sci Rep 2019, 9, 564
  96. 96. 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
  97. 97. Wang, X.; Ma, S.; Yang, B.; Huang, T.; Meng, N.; Xu, L.; Xing, Q.; Zhang, Y.; Zhang, K.; Li, Q.; Zhang, T.; Wu, J.; Yang, G. L.; Guan, F.; Wang, J. Resveratrol promotes hUC-MSCs engraftment and neural repair in a mouse model of Alzheimer's disease. Behav Brain Res 2018, 339, 297-304
  98. 98. Ross, C. A.; Akimov, S. S. Human-induced pluripotent stem cells: potential for neurodegenerative diseases. Hum Mol Genet 2014, 23, R17-R26
  99. 99. 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.; Rueda, R.; Kritskiy, O.; Abdurrob, F.; Peng, Z.; Milo, B.; Yu, C. J.; Elmsaouri, S.; Dey, D.; Ko, T.; Yankner, B. A.; Tsai, L. H. APOE4 Causes Widespread Molecular and Cellular Alterations Associated with Alzheimer's Disease Phenotypes in Human iPSC-Derived Brain Cell Types. Neuron 2018, 98, 1294
  100. 100. Oksanen, M.; Petersen, A. J.; Naumenko, N.; Puttonen, K.; Lehtonen, S.; Gubert Olive, M.; Shakirzyanova, A.; Leskela, S.; Sarajarvi, T.; Viitanen, M.; Rinne, J. O.; Hiltunen, M.; Haapasalo, A.; Giniatullin, R.; Tavi, P.; Zhang, S. C.; Kanninen, K. M.; Hamalainen, R. H.; Koistinaho, J. PSEN1 Mutant iPSC-Derived Model Reveals Severe Astrocyte Pathology in Alzheimer's Disease. Stem Cell Reports 2017, 9, 1885-1897
  101. 101. Valadez-Barba, V.; Cota-Coronado, A.; Hernandez-Perez, O. R.; Lugo-Fabres, P. H.; Padilla-Camberos, E.; Diaz, N. F.; Diaz-Martinez, N. E. iPSC for modeling neurodegenerative disorders. Regen Ther 2020, 15, 332-339
  102. 102. Wu, Y. Y.; Chiu, F. L.; Yeh, C. S.; Kuo, H. C. Opportunities and challenges for the use of induced pluripotent stem cells in modelling neurodegenerative disease. Open Biol 2019, 9, 180177
  103. 103. Machairaki, V. Human Pluripotent Stem Cells as In Vitro Models of Neurodegenerative Diseases. Adv Exp Med Biol 2020, 1195, 93-94
  104. 104. Ochalek, A.; Mihalik, B.; Avci, H. X.; Chandrasekaran, A.; Teglasi, A.; Bock, I.; Giudice, M. L.; Tancos, Z.; Molnar, K.; Laszlo, L.; Nielsen, J. E.; Holst, B.; Freude, K.; Hyttel, P.; Kobolak, J.; Dinnyes, A. Neurons derived from sporadic Alzheimer's disease iPSCs reveal elevated TAU hyperphosphorylation, increased amyloid levels, and GSK3B activation. Alzheimers Res Ther 2017, 9, 90
  105. 105. Tcw, J. Human iPSC application in Alzheimer's disease and Tau-related neurodegenerative diseases. Neurosci Lett 2019, 699, 31-40
  106. 106. Rowland, H. A.; Hooper, N. M.; Kellett, K. A. B. Modelling Sporadic Alzheimer's Disease Using Induced Pluripotent Stem Cells. Neurochem Res 2018, 43, 2179-2198
  107. 107. Sullivan, S. E.; Young-Pearse, T. L. Induced pluripotent stem cells as a discovery tool for Alzheimers disease. Brain Res 2017, 1656, 98-106
  108. 108. Adamiak, M.; Cheng, G.; Bobis-Wozowicz, S.; Zhao, L.; Kedracka-Krok, S.; Samanta, A.; Karnas, E.; Xuan, Y. T.; Skupien-Rabian, B.; Chen, X.; Jankowska, U.; Girgis, M.; Sekula, M.; Davani, A.; Lasota, S.; Vincent, R. J.; Sarna, M.; Newell, K. L.; Wang, O. L.; Dudley, N.; Madeja, Z.; Dawn, B.; Zuba-Surma, E. K. Induced Pluripotent Stem Cell (iPSC)-Derived Extracellular Vesicles Are Safer and More Effective for Cardiac Repair Than iPSCs. Circ Res 2018, 122, 296-309
  109. 109. Liu, X. Y.; Yang, L. P.; Zhao, L. Stem cell therapy for Alzheimer's disease. World J Stem Cells 2020, 12, 787-802
  110. 110. Kang, J. M.; Yeon, B. K.; Cho, S. J.; Suh, Y. H. Stem Cell Therapy for Alzheimer's Disease: A Review of Recent Clinical Trials. J Alzheimers Dis 2016, 54, 879-889
  111. 111. Yang, H.; Xie, Z.; Wei, L.; Yang, H.; Yang, S.; Zhu, Z.; Wang, P.; Zhao, C.; Bi, J. Human umbilical cord mesenchymal stem cell-derived neuron-like cells rescue memory deficits and reduce amyloid-beta deposition in an AbetaPP/PS1 transgenic mouse model. Stem Cell Res Ther 2013, 4, 76
  112. 112. Reddy, A. P.; Ravichandran, J.; Carkaci-Salli, N. Neural regeneration therapies for Alzheimer's and Parkinson's disease-related disorders. Biochim Biophys Acta Mol Basis Dis 2020, 1866, 165506

Written By

Kun Jiang, Yongqi Zhu and Lei Zhang

Submitted: 03 August 2021 Reviewed: 07 September 2021 Published: 24 November 2021