Efficacy of Adipose-Derived Stem Cells as a Potential Cellular Therapy of Alzheimer’s Disease: A Comprehensive Systematic Review

authors:

avatar Grady Krisandi ORCID 1 , 2 , avatar Rejoel Mangasa Siagian 1 , avatar Karina Karina ORCID 2 , 3 , 4 , 5 , avatar Imam Rosadi ORCID 2 , 6 , *

Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
HayandraLab, Yayasan Hayandra Peduli, Jakarta, Indonesia
Klinik Hayandra, Yayasan Hayandra Peduli, Jakarta, Indonesia
Faculty of Medicine, Universitas Pembangunan Nasional Veteran Jakarta, Indonesia
Pusat Kajian Stem Cell, Universitas Pembangunan Nasional Veteran Jakarta, Indonesia
Department of Biology, Faculty of Mathematics and Natural Sciences, Mulawarman University, Samarinda, Indonesia

how to cite: Krisandi G, Mangasa Siagian R, Karina K, Rosadi I. Efficacy of Adipose-Derived Stem Cells as a Potential Cellular Therapy of Alzheimer’s Disease: A Comprehensive Systematic Review. J Skin Stem Cell. 2020;7(4):e109466. https://doi.org/10.5812/jssc.109466.

Abstract

Context:

Elderlies are more prone to neurodegenerative diseases. One of the most common neurodegenerative diseases with limited effective treatments available is Alzheimer’s disease (AD). Adipose-derived stem cells (ADSCs) with neuroregenerative and neuroprotective capabilities have shown great potential to serve as a potential cellular therapy for AD. Adipose-derived stem cells’ efficacy in the treatment of AD is evaluated in this systematic review.

Data Sources:

Literature search was performed via PubMed, Cochrane Library, ScienceDirect, Wiley Online Library, and EBSCOhost databases, selecting studies which evaluated the effect of ADSCs on AD from inception to 6 August 2020. The SYRCLE Risk of Bias tool was used to assess pre-clinical studies in animal models, and MINORS Risk of Bias tool was used to assess non-randomized phase 1 clinical trials.

Conclusions:

Overall, seven studies, including two in-vitro studies, four in-vivo mice model studies, and one non-randomized phase 1 clinical trial, were included in this systematic review. Adipose-derived stem cells showed significant potential, proven by their neuroregenerative (i.e., increased neurogenesis, synaptogenesis, synaptic and dendritic stability, and cognitive and spatial skills) and neuroprotective capabilities (i..e., decreased Aβ concentration, APP-CT concentration, P-tau concentration, number of amyloid plaques, oxidative stress, and neuron apoptosis when exposed to Aβ). Adipose-derived stem cells demonstrated promising efficacy in treating AD patients.

1. Context

Elderlies are more susceptible to degenerative diseases including neurodegenerative diseases. The most common neurodegenerative disease among elderlies is Alzheimer’s disease (AD) (1). Alzheimer’s disease has always been a challenge in healthcare since it is the main cause of dementia, with 60 - 80% of AD patients suffering from dementia (2, 3). The incidence of AD is high and keeps rising every year (4). According to the World Alzheimer Report, there were about 40 million cases of AD in 2015. About US$ 818 billion is annually spent for research on AD, and every three seconds one new case of AS is diagnosed. The situation is further deteriorated with the 325% increase in the incidence of the disease by 2040 (5, 6).

Clinical manifestations of AD also affect the well-being of elderlies. Alzheimer’s disease may manifest with a progressive decrease in cognitive skills, language, visuospatial skills, personality, and behaviors, which may lead to the complete loss of basic daily life activities (7). These clinical manifestations are due to the pathogenesis of AD causing neurotoxicity (2, 8-11).

The pathology of AD roots in the formation of senile plaque (SP), and the accumulation of amyloid beta (Aβ) and neurofibrillary tangles (NFT) is the most common cause of SP. The formation of SP is through various cascades initiated when Aβ is present due to the mutation of the PS1/2 gene of the amyloid precursor protein (APP). This cascade involves the formation of Aβ oligomer, which causes synaptic dysfunction and neurohormonal instability. The presence of Aβ also induces the activation of inflammatory microglia and astrocytes, which play a role in local inflammation and increased oxidative stress. This leads to the alteration of kinase and phosphatase and hyperphosphorylation of tau protein (P-tau) creating NFT. Like Aβ oligomer, NFT also disturbs synaptic communication (2, 8-11).

Currently, non-pharmacological and pharmacological therapies are available. Non-pharmacological therapy utilizes various factors such as open communication between healthcare provider and patient, behavioral therapy through consistency and simplifying the environment, cognitive-behavioral therapy, light therapy, and music therapy (2, 12, 13). In pharmacological therapy, on the other hand, acetylcholinesterase inhibitors and memantine are used. However, non-pharmacological and pharmacological therapies can only maintain patient’s quality of life and are unable to reduce the severity of the disease (14).

A potential therapy for AD is utilizing adipose-derived stem cells (ADSCs), which are extracted through liposuction, a minimally invasive procedure. Besides being minimally invasive to extract, ADSCs are abundant in source because fat tissue can be found almost everywhere in the body. Another rationale for utilizing ADSCs as cellular therapy is their multipotency and autologous usage, which allows to avoid immunological rejection and tumorigenesis (15-17).

2. Objectives

This systematic review was performed to review the outcomes of treatment with adipose-derived stem cells in Alzheimer’s disease starting from in-vitro, in-vivo, and clinical trials.

3. Data Sources

3.1. Search Strategy

The preferred reporting items for systematic reviews and meta-analysis (PRISMA) was used to create this systematic review. Relevant studies were retrieved from PubMed, Cochrane Library, ScienceDirect, Wiley Online Library, and EBSCOhost from inception to 6 August 2020, with search terms as following: (“adipose tissue-derived stem cells” [All Fields] OR “stromal vascular fraction” [All Fields]) AND (“Alzheimer” [All Fields]). The search included preclinical studies and randomized controlled trials with no language restrictions applied. However, studies included in this review were restricted to English and Bahasa Indonesia, which were the only languages readable by the reviewers. Figure 1 shows the details of the literature search strategy.

Literature search strategy
Literature search strategy

3.2. Inclusion and Exclusion Criteria

The inclusion criteria for the included studies were: (1) study design: preclinical studies and clinical trials (either randomized or non-randomized); (2) language: English or Bahasa Indonesia; (3) comparison and intervention: ADSCs as main treatment; and (4) outcome parameter: Aβ peptide concentration, P-tau concentration, and subjects’ cognitive and spatial skills. Furthermore, the exclusion criteria comprised of (1) no extractable data and (2) irretrievable full-text articles.

3.3. Study Selection

The selection process for the included studies in the systematic review is shown in Figure 1. The initial search yielded 222 studies, of which 150 studies were irrelevant and 51 studies were excluded after screening the title and/or abstract. Furthermore, the excluded studies were based on no relevancy to the pre-defined inclusion criteria. Thus, seven studies were included in this systematic review, of which two were in-vitro studies, four were in-vivo studies, and one was a clinical trial.

3.4. Data Extraction and Quality Assessment

Literature search and data extraction were performed by two independent reviewers (GK and RMS), and disagreements were resolved through discussion. The following information was extracted from each study: author indicated by reference number, study design, intervention methods used, subjects, bioactive factors measured, outcomes, Aβ concentration, amyloid precursor protein C-terminal fragment (APP-CT) concentration, number of amyloid plaques, oxidative stress, bioactive factors, neprilysin (NEP) concentration, neurogenesis, synaptogenesis, synaptic and dendritic stability, neuron apoptosis, blood-brain barrier (BBB) penetration, anti-inflammatory microglial activation, and cognitive and spatial skills.

The two reviewers (GK and RMS) assessed the included studies’ quality. For the evaluation of the perceived risk of bias within the preclinical studies on animal model, the Systematic Review Centre for laboratory animal experimentation (SYRCLE) Risk of Bias tool was used (18), while the non-randomized phase 1 clinical study without control group was evaluated using the methodological item for non-randomized studies (MINORS) tool (19). No pre-determined guidelines were available to assess the bias risk for in-vitro studies.

4. Results

4.1. Characteristics of the Articles

The characteristics of the included studies are shown in Table 1. The studies mostly aimed to investigate the efficacy of ADSCs in AD with in-vitro and in-vivo studies also aiming to find the rationale behind the efficacy. In addition, two in-vivo studies investigated whether ADSCs could penetrate BBB of AD, and the non-randomized phase 1 clinical trial investigated the safety of ADSCs in 10 AD patients.

Table 1.

Study Characteristics

ReferenceStudy CharacteristicsSubject CharacteristicsIntervention
Study DesignPhaseAims/PurposeSubjectsIntervention Treatment GroupsIntervention MethodsBioactive Factors
(20)In-vitro-To examine efficacy of ADSC-Exo in ADNeutrosphere cells from TG2576 transgenic micehADSC72 hours co-cultureADSC-Exo
(21)In-vitro-To examine efficacy of ADSC-Exo (NEP) in ADN2a cell linehADSC56 hours co-cultureADSC-Exo (NEP)
(22)In-vivo-To examine the efficacy of ADSC in activating the alternative phenotype microglia of ADAPP/PS1 transgenic micemADSCIntracerebral injectionIL-4 and Arginase I
(23)In-vivo-To examine the beneficial effects and neurogenesis of ADSC in ADAPP/PS1 transgenic micemADSCIntrahippocampal injectionADSC-Exo
(24)In-vivo-To examine if ADSC could penetrate BBB; efficacy of ADSC in the early (intracerebral injection) and late stage (intravenous injection) of AD; efficacy of ADSC in preventing ADAPPswe TG2576 transgenic micehADSCIntracerebral injection; intravenous injectionADSC-Exo (NEP), IL-10, and VEGF
(25)In-vivo-To examine if ADSC could penetrate BBBTG2576 transgenic micehADSCIntravenous injection-
(26)Non-randomized clinical trialITo examine safety and efficacy of ADSC in AD patients10 Alzheimer patientsSVFSubgaleal ommaya reservoir-

4.2. Risk of Bias within Studies

Pre-clinical studies in animal model were assessed using the SYRCLE Risk of Bias tool. As for the preclinical in-vivo studies on animal models, four preclinical studies were rated on the perceived risk of bias using SYRCLE tool. For two of them, the allocation sequence was reported unclearly, raising concern for selection bias. Moreover, all the studies were not documented for blinding housing, blinding intervention, and blinding outcome assessment. These would raise some concern regarding the potential for performance bias and detection bias. Lastly, the included in-vivo studies were free of risk of reporting and other biases. The summary of risk of bias for preclinical studies in animal models is presented in Table 2.

Table 2.

Risk of Bias Assessment for In-vivo Studies a

Type of Bias and ItemsSYRCLE’s Tool for Assessing Risk of Bias (18)
DomainQuestionReference
(22)(24)(25)(23)
Selection bias
1Sequence generationWas the allocation sequence adequately generated and applied?YesYesUnclearUnclear
2Baseline characteristicsWere the groups similar at baseline or were they adjusted for confounders in the analysis?YesYesUnclearUnclear
3Allocation concealmentWas the allocation adequately concealed?NoYesYesYes
Performance bias
4Random housingWere the randomly animals housed along the experiment?UnclearNoUnclearUnclear
5BlindingWere the caregivers and /or investigators blinded from knowledge which intervention each animal received during the experiment?NoUnclearUnclearNo
Detection bias
6Random outcome assessmentWere animals selected at random for outcome assessment?UnclearNoUnclearNo
7BlindingWas the outcome assessor blinded?UnclearNoNoNo
Attrition bias
8Incomplete outcome dataWere incomplete outcome data adequately addressed?YesYesNoYes
Reporting bias
9Selective outcome reportingWas the study free of selective outcome?YesYesYesYes
Other
10Bias from another sourcesIs the study free of problems leads to high risk of bias?YesYesYesYes

Assessment of the perceived risk of bias in the non-randomized phase 1 clinical study was performed using the MINORS Risk of Bias tool. The study did not apply a control group and was non-randomized; thus, there was no comparison or blinding. This explains the 0 score for items 8 - 11 (Table 3). It raises concern regarding the potential bias of the outcome report. Lastly, the study reported adequate statistical analysis to support the outcome documented in the study. The summary of risk of bias for the non-randomized phase 1 clinical study is shown in Table 3.

Table 3.

Risk of Bias Assessment for Non-randomized Clinical Trial Study a

MINORS tool for assessing risk of bias (19)
Item ComponentsDuma et al. [2019] (26)
1A purpose of study2
2Patient inclusion2
3Data collection2
4Goal of study related to the purpose2
5Assessment of study unbiased2
6Period of patient follow-up2
7Losing of patient follow up (< 5%)2
8The size study size0
9Group of control0
10Contemporary groups0
11Groups baseline0
12Analysis of statistics1
Total Score15

4.3. Efficacy

Effects of ADSCs on AD (Table 4) were evaluated by two in-vitro studies, four in-vivo mouse model studies, and one clinical trial. Outcomes extracted for assessment include Aβ concentration, P-tau concentration, APP-CT concentration, number of amyloid plaques, oxidative stress, bioactive factors, NEP concentration, neurogenesis, synaptogenesis, synaptic and dendritic stability, neuron apoptosis, BBB penetration, activation of alternative phenotype microglia, cognitive skills, and spatial skills.

Table 4.

Reported Outcomes Related to the Efficacy of ADSCs in AD

ReferenceOutcomes
Aβ ConcentrationP-Tau ConcentrationAPP-CT ConcentrationNumber of Amyloid PlaquesOxidative StressBioactive FactorsNeprilysin ConcentrationNeurogenesisSynaptogenesisSynaptic And Dendritic StabilityNeuron ApoptosisBlood-Brain Barrier PenetrationAlternative Phenotype MicrogliaCognitive SkillsSpatial Skills
(20)Decreased (P < 0.01)------Increased (P < 0.01)Increased (P < 0.01)-Decreased (P < 0.01)----
(21)Decreased (intracellular and extracellular) (P < 0.001)--------------
(22)Decreased (hippocampus: P < 0.01; cortex: P > 0.05)--Decreased (hippocampus: P < 0.01; cortex: P < 0.05)-Decreased mRNA expression of TNF-α (hippocampus: P < 0.01; cortex: P < 0.05) and IL-1β (hippocampus: P < 0.01; cortex: P < 0.05); increased mRNA expression of IL-4 (hippocampus and cortex: P < 0.01) and ARG1 (hippocampus and cortex: P < 0.01); increased mRNA expression of IDE (hippocampus and cortex: P < 0.01) and MMP-9 (hippocampus: P < 0.01; cortex: P > 0.05)Increased (P < 0.01)-----Activated (P < 0.01)Increased (P < 0.05)-
(23)----Decreased (P < 0.01) (hippocampus)--Increased (P < 0.01)-----Increased (P < 0.05)-
(24)Decreased (IC: P < 0.05; IV: P < 0.05)-Decreased (IC: P < 0.05; IV: P < 0.05)Decreased (IC/cortex: P < 0.05; IC/hippocampus: P < 0.05; IV/cortex: P < 0.05; IV/hippocampus: P < 0.14)-Increased IL-10 (IC and IV: P < 0.05) and VEGF (IC 6 weeks: P < 0.01; IC and IV 4 months: P < 0.05)Increased (IC: 1.29-fold, P < 0.05; IV: 1.14-fold, P < 0.05)Increased (IC: P < 0.05; IV: P > 0.05)-IncreasedDecreased (P < 0.05)PenetratedActivated-Increased (IC: P < 0.01; IV: P < 0.05)
(25)-----------Penetrated---
(26)-Decreased (3 patients)-----Increased (2 patients had an increase of hippocampal volume)-----Increased (5 patients)-

Overall, in-vitro and in-vivo studies showed a significant reduction in Aβ concentration, APP-CT concentration, number of amyloid plaques, oxidative stress, neuron apoptosis when exposed to Aβ, and mRNA expression of TNF-α and IL-1β. A fa fall in P-tau was observed in three patients in the non-randomized phase 1 clinical trial. On the other hand, in-vitro and in-vivo studies exhibited a significant increase in IL-4 mRNA expression, IL-10, vascular endothelial growth factors (VEGF), and NEP production, neurogenesis, synaptogenesis, synaptic and dendritic stability, cognitive skills, and spatial skills. The non-randomized phase 1 clinical trial also showed an improvement in cognitive skills in five patients and an increase in hippocampal volume in two patients. An intriguing finding was the ability of ADSCs to penetrate the BBB of AD patients and activate the alternative phenotype microglia.

5. Discussion

The regenerative potential of ADSCs has been studied and used for osteochondral tissue engineering and cell therapies, such as wound healing and calvarial defects (27). Based on the systematic review, we found that ADSCs also have a potential to be used in cellular therapy for AD.

The potential of ADSCs to be used as a regenerative cellular therapy for AD lies in their multipotency and autologous usage. This enables ADSCs to create any tissue in the body, except placenta, and avoid immunologic rejection. The differentiation of ADSCs is usually induced when they receive certain signals from damaged tissue that make them acquire tissue-specific phenotypes. Aside from pluripotency, the regenerative effects of ADSCs lie mainly in their autocrine production of various bioactive factors, such as growth factors, cytokines, chemokines, antiapoptotic factors, and immunomodulators in the form of gene products such as mRNA and miRNA species (17). These bioactive factors are often secreted in the form of exosomes and may affect the behavior of affected cells. Furthermore, bioactive factors which have immunomodulatory effects result in positive effects on AD, which is worsened through neuroinflammation in the CNS (17, 28).

The reviewed studies showed that ADSCs increased neurogenesis, synaptogenesis, and the stability of synapses and dendrites. The rationale behind this finding mostly lies in the multipotency of ADSCs, exosomes produced, and their ability to penetrate the BBB. Differentiation ADSCs can be induced by reactive oxygen species (ROS). In AD, excessive ROS induced by elevated levels of Aβ peptide leads to oxidative stress and inappropriate microenvironment for neurogenesis. Although ROS can induce differentiation of neural stem cells (NSC) or ADSCs, oxidative stress results in toxicity and cell death of the newly differentiated neurons. Interestingly, ADSCs have the ability to reduce ROS and increase the self-renewal capacity of the CNS that leads to neurogenesis through NADPH oxidase and PI3K/Akt pathway (23, 29). The possible mechanism behind ADSCs reducing ROS is through the exosomes produced, which may decrease ROS generation, enhance ROS’ scavenging capability, or repair oxidized molecules (23). Furthermore, reduced ROS also explains that increased neurogenesis promotes synaptogenesis and prevents neuronal membrane damage, which leads to the enhanced stability of both synapses and dendrites (30).

Aside from ROS reduction, exosomes of ADSCs (ADSC-Exo) have been found to induce remyelination in damaged neurons, which also explains the neuroregenerative capability of ADSCs (31). Moreover, ADSC-Exo have been reported to upregulate VEGF.26 It has been reported that VEGF plays a role in neuronal wiring of the CNS by regulating neuronal cell migration and neuroregeneration, which explains the increased neurogenesis, synaptogenesis, and stability of synapses and dendrites (32).

The ability of ADSCs to reduce ROS in AD also explains the reduced neuron apoptosis and increased neuron viability observed (24). Reports on the upregulation of VEGF by ADSC-Exo also explain the decrease in neuron apoptosis and increase in neuron viability. Vascular endothelial growth factors are reported to be able to inhibit Aβ-induced cytotoxicity in neurons. In AD, amyloid plaques co-aggregate with the VEGF produced. This causes VEGF depletion and neurodegeneration due to reduced neuroprotective effects (32). This phenomenon is further explained by the ability of ADSC-Exo to exert their neuroprotective effects against glutamate excitotoxicity, which causes neurodegeneration in AD. The mechanism through which ADSC-Exo exert their neuroprotective effects is the amelioration of neural recovery marker. The neural recovery marker ameliorated is the growth-associated protein 43 (GAP-43) and the number of GAP-43 positive neurites, which leads to neural development and regeneration. Another mechanism of ADSC-Exo is through increasing the levels of ATP, NAD, and NADH, which rescues neurons from glutamate-induced neuronal energy depletion in AD (33-35).

The reviewed studies also reported the activation of the alternative phenotypes of microglia by ADSCs. These studies reported this finding through the observation of strong expression of IL-4 and Arg-1, which are the markers of alternative phenotype microglia. This activation is beneficial because they play a protective role against AD by mediating the clearance of Aβ and reducing the production of proinflammatory and neurotoxic cytokines, which does not worsen the pathological course of AD. the clearance of Aβ is mediated through the production of IL-4 and IL-10, which are neuroprotective and through the phagocytosis of Aβ by alternative phenotype microglia (36). The production of IL-4 by alternative phenotype microglia further activates alternative phenotype microglia (37). On the other hand, the production of IL-10 is due to its ability in neutralizing cytotoxic inflammatory processes induced by Aβ and down-regulating IL-1β and TNF-α synthesis (38). The down-regulation of pro-inflammatory cytokines is beneficial towards the phagocytic ability of alternative phenotype microglia, which is attenuated by pro-inflammatory cytokines (39).

Furthermore, the degradation of Aβ is not only by phagocytosis through microglia, but ADSC-Exo also play a role in the degradation of Aβ. It has been found that ADSC-Exo stimulate increased production of Aβ degradative enzymes. These enzymes include insulin-degrading enzyme (IDE) and NEP (40, 41). Although the two of them are able to degrade Aβ both intracellularly and extracellularly, the degradative mechanisms differ between the two of them. Neprilysin degrades Aβ by cleaving peptides on the N-terminal of hydrophobic residues and has broad substrate specificity (42, 43). On the contrary, IDE is only proteolytic towards β-structure-forming substrates, which makes them the major protease responsible for Aβ clearance (41). Moreover, an increase in matrix metalloproteinase-9 (MMP-9) was also reported and may also explain the reduction of Aβ. Studies have found that MMP-9 exerts neuroprotective effects through the cleavage of APP by enhancing α-secretase activity. With the enhanced cleavage of APP, the production of Aβ will be diminished (43). Undoubtedly, the decrease in Aβ will cause reduce ROS and will also stop the formation of both P-tau and NFT, which explains a reported finding in this review (11).

Lastly, studies reviewed also reported that ADSCs alleviate cognitive and spatial skills impairment. The main cause of cognitive and spatial skills impairment is the assembly of Aβ, which causes many pathological effects (44). Adipose-derived stem cells’ ability to degrade Aβ explains this finding (40, 41).

5.1. Conclusions

In light of all the evidence presented in this systematic review, ADSCs have an excellent potential as a novel therapy for AD. Preclinical studies found that ADSCs have neuroregenerative and neuroprotective capabilities and reduce the concentration of Aβ, which plays a main role in various pathological processes in the progression of AD. These findings were further proven in another preclinical study and a non-randomized phase 1 clinical trial, which showed that ADSCs could alleviate cognitive and spatial skills impairment.

References

  • 1.

    Bondi MW, Edmonds EC, Salmon DP. Alzheimer's Disease: Past, Present, and Future. J Int Neuropsychol Soc. 2017;23(9-10):818-31. [PubMed ID: 29198280]. [PubMed Central ID: PMC5830188]. https://doi.org/10.1017/S135561771700100X.

  • 2.

    Yiannopoulou KG, Papageorgiou SG. Current and Future Treatments in Alzheimer Disease: An Update. J Cent Nerv Syst Dis. 2020;12:1179573520907400. [PubMed ID: 32165850]. [PubMed Central ID: PMC7050025]. https://doi.org/10.1177/1179573520907397.

  • 3.

    Alzheimer's Association. 2018 Alzheimer's disease facts and figures. Alzheimer's & Dementia. 2018;14(3):367-429. https://doi.org/10.1016/j.jalz.2018.02.001.

  • 4.

    Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimers Dement. 2013;9(1):63-75 e2. [PubMed ID: 23305823]. https://doi.org/10.1016/j.jalz.2012.11.007.

  • 5.

    Prince MJ, Comas -A, Knapp M, Guerchet MM, Karagiannidou M. World Alzheimer report 2016: improving healthcare for people living with dementia: coverage, quality and costs now and in the future. London (UK): Alzheimer's Disease International; 2016.

  • 6.

    Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease. Nat Rev Neurol. 2011;7(3):137-52. [PubMed ID: 21304480]. [PubMed Central ID: PMC3339565]. https://doi.org/10.1038/nrneurol.2011.2.

  • 7.

    Weller J, Budson A. Current understanding of Alzheimer's disease diagnosis and treatment. F1000Res. 2018;7. [PubMed ID: 30135715]. [PubMed Central ID: PMC6073093]. https://doi.org/10.12688/f1000research.14506.1.

  • 8.

    Fan L, Mao C, Hu X, Zhang S, Yang Z, Hu Z, et al. New Insights Into the Pathogenesis of Alzheimer's Disease. Front Neurol. 2019;10:1312. [PubMed ID: 31998208]. [PubMed Central ID: PMC6965067]. https://doi.org/10.3389/fneur.2019.01312.

  • 9.

    Yiannopoulou KG, Papageorgiou SG. Current and future treatments for Alzheimer's disease. Ther Adv Neurol Disord. 2013;6(1):19-33. [PubMed ID: 23277790]. [PubMed Central ID: PMC3526946]. https://doi.org/10.1177/1756285612461679.

  • 10.

    Kaushal V, Dye R, Pakavathkumar P, Foveau B, Flores J, Hyman B, et al. Neuronal NLRP1 inflammasome activation of Caspase-1 coordinately regulates inflammatory interleukin-1-beta production and axonal degeneration-associated Caspase-6 activation. Cell Death Differ. 2015;22(10):1676-86. [PubMed ID: 25744023]. [PubMed Central ID: PMC4563782]. https://doi.org/10.1038/cdd.2015.16.

  • 11.

    Tiwari S, Atluri V, Kaushik A, Yndart A, Nair M. Alzheimer's disease: pathogenesis, diagnostics, and therapeutics. Int J Nanomedicine. 2019;14:5541-54. [PubMed ID: 31410002]. [PubMed Central ID: PMC6650620]. https://doi.org/10.2147/IJN.S200490.

  • 12.

    Atri A. Current and future treatments in alzheimer's disease. Semin Neurol. 2019;39(2):227-40. [PubMed ID: 30925615]. https://doi.org/10.1055/s-0039-1678581.

  • 13.

    Kishita N, Backhouse T, Mioshi E. Nonpharmacological Interventions to Improve Depression, Anxiety, and Quality of Life (QoL) in People With Dementia: An Overview of Systematic Reviews. J Geriatr Psychiatry Neurol. 2020;33(1):28-41. [PubMed ID: 31203712]. https://doi.org/10.1177/0891988719856690.

  • 14.

    Mossello E, Ballini E. Management of patients with Alzheimer's disease: pharmacological treatment and quality of life. Ther Adv Chronic Dis. 2012;3(4):183-93. [PubMed ID: 23342234]. [PubMed Central ID: PMC3539289]. https://doi.org/10.1177/2040622312452387.

  • 15.

    Karina K, Rosliana I, Rosadi I, Schwartz R, Sobariah S, Afini I, et al. Safety of technique and procedure of stromal vascular fraction therapy: From liposuction to cell administration. Scientifica (Cairo). 2020;2020:2863624. [PubMed ID: 32695550]. [PubMed Central ID: PMC7362293]. https://doi.org/10.1155/2020/2863624.

  • 16.

    Hong P, Yang H, Wu Y, Li K, Tang Z. The functions and clinical application potential of exosomes derived from adipose mesenchymal stem cells: a comprehensive review. Stem Cell Res Ther. 2019;10(1):242. [PubMed ID: 31391108]. [PubMed Central ID: PMC6686455]. https://doi.org/10.1186/s13287-019-1358-y.

  • 17.

    Bacakova L, Zarubova J, Travnickova M, Musilkova J, Pajorova J, Slepicka P, et al. Stem cells: their source, potency and use in regenerative therapies with focus on adipose-derived stem cells - a review. Biotechnol Adv. 2018;36(4):1111-26. [PubMed ID: 29563048]. https://doi.org/10.1016/j.biotechadv.2018.03.011.

  • 18.

    Hooijmans CR, Rovers MM, de Vries RB, Leenaars M, Ritskes-Hoitinga M, Langendam MW. SYRCLE's risk of bias tool for animal studies. BMC Med Res Methodol. 2014;14:43. [PubMed ID: 24667063]. [PubMed Central ID: PMC4230647]. https://doi.org/10.1186/1471-2288-14-43.

  • 19.

    Slim K, Nini E, Forestier D, Kwiatkowski F, Panis Y, Chipponi J. Methodological index for non-randomized studies (minors): development and validation of a new instrument. ANZ J Surg. 2003;73(9):712-6. [PubMed ID: 12956787]. https://doi.org/10.1046/j.1445-2197.2003.02748.x.

  • 20.

    Lee M, Ban JJ, 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. [PubMed ID: 29625119]. https://doi.org/10.1016/j.brainres.2018.03.034.

  • 21.

    Katsuda T, Tsuchiya R, Kosaka N, Yoshioka Y, Takagaki K, Oki K, et al. Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Sci Rep. 2013;3:1197. [PubMed ID: 23378928]. [PubMed Central ID: PMC3561625]. https://doi.org/10.1038/srep01197.

  • 22.

    Ma T, Gong K, Ao Q, Yan Y, Song B, Huang H, et al. Intracerebral transplantation of adipose-derived mesenchymal stem cells alternatively activates microglia and ameliorates neuropathological deficits in Alzheimer's disease mice. Cell Transplant. 2013;22 Suppl 1:S113-26. [PubMed ID: 24070198]. https://doi.org/10.3727/096368913X672181.

  • 23.

    Yan Y, Ma T, Gong K, Ao Q, Zhang X, Gong Y. Adipose-derived mesenchymal stem cell transplantation promotes adult neurogenesis in the brains of Alzheimer's disease mice. Neural Regen Res. 2014;9(8):798-805. [PubMed ID: 25206892]. [PubMed Central ID: PMC4146257]. https://doi.org/10.4103/1673-5374.131596.

  • 24.

    Kim S, Chang KA, Kim J, Park HG, Ra JC, Kim HS, et al. The preventive and therapeutic effects of intravenous human adipose-derived stem cells in Alzheimer's disease mice. PLoS One. 2012;7(9). e45757. [PubMed ID: 23049854]. [PubMed Central ID: PMC3458942]. https://doi.org/10.1371/journal.pone.0045757.

  • 25.

    Ha S, Ahn S, Kim S, Joo Y, Chong YH, Suh YH, et al. In vivo imaging of human adipose-derived stem cells in Alzheimer's disease animal model. J Biomed Opt. 2014;19(5):51206. [PubMed ID: 24297061]. https://doi.org/10.1117/1.JBO.19.5.051206.

  • 26.

    Duma C, Kopyov O, Kopyov A, Berman M, Lander E, Elam M, et al. Human intracerebroventricular (ICV) injection of autologous, non-engineered, adipose-derived stromal vascular fraction (ADSVF) for neurodegenerative disorders: results of a 3-year phase 1 study of 113 injections in 31 patients. Mol Biol Rep. 2019;46(5):5257-72. [PubMed ID: 31327120]. https://doi.org/10.1007/s11033-019-04983-5.

  • 27.

    Tsuji W, Rubin JP, Marra KG. Adipose-derived stem cells: Implications in tissue regeneration. World J Stem Cells. 2014;6(3):312-21. [PubMed ID: 25126381]. [PubMed Central ID: PMC4131273]. https://doi.org/10.4252/wjsc.v6.i3.312.

  • 28.

    Mammana S, Fagone P, Cavalli E, Basile MS, Petralia MC, Nicoletti F, et al. The role of macrophages in neuroinflammatory and neurodegenerative pathways of alzheimer's disease, amyotrophic lateral sclerosis, and multiple sclerosis: Pathogenetic cellular effectors and potential therapeutic targets. Int J Mol Sci. 2018;19(3). [PubMed ID: 29533975]. [PubMed Central ID: PMC5877692]. https://doi.org/10.3390/ijms19030831.

  • 29.

    Le Belle JE, Orozco NM, Paucar AA, Saxe JP, Mottahedeh J, Pyle AD, et al. Proliferative neural stem cells have high endogenous ROS levels that regulate self-renewal and neurogenesis in a PI3K/Akt-dependant manner. Cell Stem Cell. 2011;8(1):59-71. [PubMed ID: 21211782]. [PubMed Central ID: PMC3018289]. https://doi.org/10.1016/j.stem.2010.11.028.

  • 30.

    Farooqui T, Farooqui AA. Aging: an important factor for the pathogenesis of neurodegenerative diseases. Mech Ageing Dev. 2009;130(4):203-15. [PubMed ID: 19071157]. https://doi.org/10.1016/j.mad.2008.11.006.

  • 31.

    Farinazzo A, Turano E, Marconi S, Bistaffa E, Bazzoli E, Bonetti B. Murine adipose-derived mesenchymal stromal cell vesicles: in vitro clues for neuroprotective and neuroregenerative approaches. Cytotherapy. 2015;17(5):571-8. [PubMed ID: 25743633]. https://doi.org/10.1016/j.jcyt.2015.01.005.

  • 32.

    Ruiz de Almodovar C, Lambrechts D, Mazzone M, Carmeliet P. Role and therapeutic potential of VEGF in the nervous system. Physiol Rev. 2009;89(2):607-48. [PubMed ID: 19342615]. https://doi.org/10.1152/physrev.00031.2008.

  • 33.

    Cai Y, Li J, Jia C, He Y, Deng C. Therapeutic applications of adipose cell-free derivatives: a review. Stem Cell Res Ther. 2020;11(1):312. [PubMed ID: 32698868]. [PubMed Central ID: PMC7374967]. https://doi.org/10.1186/s13287-020-01831-3.

  • 34.

    Hao P, Liang Z, Piao H, Ji X, Wang Y, Liu Y, et al. Conditioned medium of human adipose-derived mesenchymal stem cells mediates protection in neurons following glutamate excitotoxicity by regulating energy metabolism and GAP-43 expression. Metab Brain Dis. 2014;29(1):193-205. [PubMed ID: 24458787]. [PubMed Central ID: PMC3930846]. https://doi.org/10.1007/s11011-014-9490-y.

  • 35.

    Hynd MR, Scott HL, Dodd PR. Glutamate-mediated excitotoxicity and neurodegeneration in Alzheimer's disease. Neurochem Int. 2004;45(5):583-95. [PubMed ID: 15234100]. https://doi.org/10.1016/j.neuint.2004.03.007.

  • 36.

    Lee JK, Jin HK, Endo S, Schuchman EH, Carter JE, Bae JS. Intracerebral transplantation of bone marrow-derived mesenchymal stem cells reduces amyloid-beta deposition and rescues memory deficits in Alzheimer's disease mice by modulation of immune responses. Stem Cells. 2010;28(2):329-43. [PubMed ID: 20014009]. https://doi.org/10.1002/stem.277.

  • 37.

    Latta CH, Sudduth TL, Weekman EM, Brothers HM, Abner EL, Popa GJ, et al. Determining the role of IL-4 induced neuroinflammation in microglial activity and amyloid-beta using BV2 microglial cells and APP/PS1 transgenic mice. J Neuroinflammation. 2015;12:41. [PubMed ID: 25885682]. [PubMed Central ID: PMC4350455]. https://doi.org/10.1186/s12974-015-0243-6.

  • 38.

    de Godoy MA, Saraiva LM, de Carvalho LRP, Vasconcelos-Dos-Santos A, Beiral HJV, Ramos AB, et al. Mesenchymal stem cells and cell-derived extracellular vesicles protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-beta oligomers. J Biol Chem. 2018;293(6):1957-75. [PubMed ID: 29284679]. [PubMed Central ID: PMC5808759]. https://doi.org/10.1074/jbc.M117.807180.

  • 39.

    Tang Y, Le W. Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol Neurobiol. 2016;53(2):1181-94. [PubMed ID: 25598354]. https://doi.org/10.1007/s12035-014-9070-5.

  • 40.

    Zhang H, Liu D, Wang Y, Huang H, Zhao Y, Zhou H. Meta-analysis of expression and function of neprilysin in Alzheimer's disease. Neurosci Lett. 2017;657:69-76. [PubMed ID: 28778804]. https://doi.org/10.1016/j.neulet.2017.07.060.

  • 41.

    Kurochkin IV, Guarnera E, Berezovsky IN. Insulin-degrading enzyme in the fight against alzheimer's disease. Trends Pharmacol Sci. 2018;39(1):49-58. [PubMed ID: 29132916]. https://doi.org/10.1016/j.tips.2017.10.008.

  • 42.

    Klein C, Roussel G, Brun S, Rusu C, Patte-Mensah C, Maitre M, et al. 5-HIAA induces neprilysin to ameliorate pathophysiology and symptoms in a mouse model for Alzheimer's disease. Acta Neuropathol Commun. 2018;6(1):136. [PubMed ID: 30537985]. [PubMed Central ID: PMC6290545]. https://doi.org/10.1186/s40478-018-0640-z.

  • 43.

    Kaminari A, Tsilibary EC, Tzinia A. A new perspective in utilizing MMP-9 as a therapeutic target for alzheimer's disease and type 2 diabetes mellitus. J Alzheimers Dis. 2018;64(1):1-16. [PubMed ID: 29865065]. https://doi.org/10.3233/JAD-180035.

  • 44.

    Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CG, Yang A, et al. A specific amyloid-beta protein assembly in the brain impairs memory. Nature. 2006;440(7082):352-7. [PubMed ID: 16541076]. https://doi.org/10.1038/nature04533.