Next Article in Journal
Piezo-Type Mechanosensitive Ion Channel Component 1 (PIEZO1) as a Potential Prognostic Marker in Renal Clear Cell Carcinoma
Previous Article in Journal
Valproic Acid Reduces Invasiveness and Cellular Growth in 2D and 3D Glioblastoma Cell Lines
Previous Article in Special Issue
Unraveling the Roles of UBE3A in Neurodevelopment and Neurodegeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

From Adipose to Action: Reprogramming Stem Cells for Functional Neural Progenitors for Neural Regenerative Therapy

1
Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China
2
Golden Meditech Centre for NeuroRegeneration Sciences, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China
3
Mr. & Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China
4
Department of Science and Environmental Studies, The Education University of Hong Kong, Tai Po, Hong Kong SAR, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(14), 6599; https://doi.org/10.3390/ijms26146599
Submission received: 4 June 2025 / Revised: 28 June 2025 / Accepted: 8 July 2025 / Published: 9 July 2025
(This article belongs to the Special Issue Challenges and Innovation in Neurodegenerative Diseases, 2nd Edition)

Abstract

Neural stem cells have shown great potential in the therapy of neurodegenerative diseases such as Parkinson’s disease (PD), because of their ability to differentiate into various types of neural cells and substitute for damaged neurons. Their clinical application is, however, impeded by limitations such as low survival rates following transplantation, low efficiency of differentiation, the potential for tumorigenesis, and the risk of immune rejection by the host. Adipose-derived stem cells (ADSCs) have become increasingly popular as an alternative tool in regenerative medicine due to their accessibility, multipotency, and low immunogenicity. The recent advance in inducing ADSCs into neural stem cell-like cells (iNSCs) opens up a new avenue for the treatment of PD by restoring dopaminergic neuron populations. Here, the biological characteristics, induction protocols, molecular mechanisms, and prospective applications of ADSCs in neural repair are summarized systematically. We also covered current technical challenges, such as differentiation protocol optimization and functional integration, and future perspectives, including biomaterial and gene editing applications to enhance ADSC-based therapies. With these challenges met, ADSCs hold excellent potential for advancing personalized and combination therapies for neurodegenerative diseases.

Graphical Abstract

1. Introduction

Neurodegenerative diseases are a group of disorders characterized by the progressive degeneration of the structure and function of the nervous system. These conditions often lead to a decline in cognitive and motor functions, significantly impacting patients’ quality of life [1]. Among these disorders, Parkinson’s disease (PD) is the second most common neurodegenerative disorder. It is characterized by the gradual degeneration of dopaminergic neurons in the substantia nigra [2,3]. Existing treatments, such as dopamine replacement therapy and deep brain stimulation, ameliorate symptoms but do not stop disease progression or restore lost dopaminergic neurons [4]. Stem cell therapy is one of the promising strategies for replenishing dopaminergic neurons and repairing the disrupted neural circuit [5,6]. But the clinical use of conventional stem cell sources, including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), is restricted due to ethical issues, tumorigenic potential, and immune rejection [7,8,9].
Adipose-derived stem cells (ADSCs) have been suggested as an alternative source of stem cells because of their accessibility, multipotency, and low immunogenicity [10,11]. In contrast to ESCs and iPSCs, ADSCs can be readily obtained from adipose tissue through less invasive procedures [12,13], making them an attractive candidate for autologous applications. Recently, autologous ADSC injections have gained popularity for treating central nervous system diseases. They can differentiate into neural stem cell-like cells (iNSCs) [14,15], suggesting a new strategy for the treatment of neurodegenerative disease, such as PD. Nonetheless, the molecular basis of ADSC transdifferentiation and their clinical use has not been well defined.
This review focuses on the biological properties, inducing methods, and molecular mechanisms of ADSCs differentiation into iNSCs. We also explore the therapeutic potential of ADSCs in neural regeneration. Additionally, we discuss existing technical challenges and future directions for optimizing ADSC-based therapies for PD. Most importantly, these challenges should be addressed as ADSCs have potential in progressing regenerative medicine and improving outcomes for neurodegenerative disease patients.

2. Characteristics of NSCs and ADSCs

Neural Stem Cells (NSCs) and ADSCs are both valuable tools in regenerative medicine because of their high capacity for self-renewal, multipotency for differentiation into different cell lineages, and ability to secrete paracrine factors [16]. However, they possess distinct biological features and therapeutic potentials. Below, we compare NSCs and ADSCs in terms of their sources, morphology, surface markers, and functional properties (Table 1).

2.1. Sources

2.1.1. NSCs

The adult brain and embryonic tissues are the major sources of NSCs. In adults, NSCs are found in specific areas like the subventricular zone (SVZ) and the dentate gyrus of the hippocampal region [17,18]. NSCs can proliferate and differentiate into neurons, astrocytes, and oligodendrocytes after being isolated, which aids in neural plasticity and nervous system repair [16,19]. Microglia are not a typical differentiation product of NSCs because they originate from yolk sac-derived primitive myeloid progenitors [20]. Embryonic NSCs, on the other hand, originate from the neural tube during early development and exhibit a strong capacity of differentiation [18,21]. However, because of ethical issues and technical difficulties in their isolation and expansion, the use of embryonic NSCs is limited [22].

2.1.2. ADSCs

ADSCs are mainly derived from the huge reserves of subcutaneous adipose [23,24]. They are typically harvested through less invasive techniques such as liposuction in humans or surgical extraction in experimental animals [25]. The harvested adipose tissue is enzymatically digested and processed to yield a cell suspension enriched with ADSCs. Notably, the typical yield of one gram of adipose tissue can be up to around 0.5 × 104 to 2 × 105 stem cells, significantly higher than the number of mesenchymal stem cells (MSCs; typically around 60–600 cells per 1 mL) obtained from bone marrow aspirate [26]. This abundance as well as the ease of isolation makes ADSCs an attractive choice for autologous transplantation and regenerative therapies [27,28].

2.2. Morphological Characteristics

2.2.1. NSCs

In vitro, NSCs typically form neurospheres—free-floating clusters of cells—when cultured in specific media [29]. These neurospheres exhibit a multi-protrusion morphology (Figure 1A). Forming neurospheres is a hallmark feature of NSCs and reflects their ability of self-renewal and multipotency [30,31].

2.2.2. ADSCs

ADSCs usually show a fibroblast-like, spindle-shaped morphology (Figure 1B) under standard culture conditions [32,33]. In contrast to the NSCs, ADSCs do not form neurospheres but they grow as an adherent monolayer instead (Figure 1B). While ADSCs share some morphological similarities with NSCs, their differentiation potential is primarily restricted to mesodermal lineages, like adipocytes, osteoblasts, chondrocytes, etc., unless specific induction protocols are applied [34,35].

2.3. Cell Surface Markers

2.3.1. NSCs

NSCs express a range of specific surface markers. These markers include Nestin, Sox2, Musashi-1 and CD133, with each having its unique function [36]. Nestin, an intermediate filament protein, is an early marker of neural progenitor cells [37]. Sox2 is a transcription factor which plays a crucial role in maintaining the self-renewal and differentiation potential of NSCs [38]. Musashi-1 is an RNA-binding protein which regulates NSC proliferation and differentiation [39], while CD133 is a transmembrane protein that is associated with stem cell identity [40].

2.3.2. ADSCs

ADSCs express mesenchymal stem cell markers such as CD29, CD90, and CD105 [41]. CD29, also known as integrin β1, mediates the adhesion of ADSCs to the extracellular matrix (such as collagen and laminin). It also regulates stem cell homing and differentiation [42]. CD90 (Thy-1) influences ADSC proliferation and differentiation through pathways such as Wnt/β-catenin [43], while CD105 (Endoglin) is involved in angiogenesis and tissue repair via the TGF-β/SMAD signaling pathway [44]. Importantly, ADSCs do not express hematopoietic makers like CD34 and CD45, ensuring their purity and suitability for clinical applications [45].

2.4. Functional Properties

2.4.1. NSCs

NSCs possess distinctive capacity for self-renewal and differentiate into diverse neural cell types, including neurons, astrocytes, and oligodendrocytes [46]. Their multipotency arises from two division mechanisms: symmetric division produces two identical NSCs, whereas asymmetric division generates one NSC and one specialized cell [47]. These cells are essential for neural repair and regeneration processes, positioning them as promising therapeutic candidates for neurodegenerative diseases [48,49].

2.4.2. ADSCs

ADSCs demonstrate exceptional multipotency, differentiating into multiple cell lineages such as osteoblasts, chondrocytes, adipocytes, hepatic lineage, and even neural-like cells under specific induction conditions [50,51]. Besides differentiation capabilities, ADSCs also secrete several neurotrophic factors, including the nerve growth factor (NGF), the brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), the ciliary neurotrophic factor (CNTF), the glial cell-derived neurotrophic factor (GDNF), and artemin [41,52,53]. These bioactive molecules induce neural repair, minimize inflammation responses, and augment tissue regeneration [54]. Moreover, ADSCs release extracellular vesicles (EVs) that carry anti-inflammatory and neuroprotective effects, suggesting their potential application in cell-free therapy approaches [55,56].
Table 1. Comparison of NSCs and ADSCs.
Table 1. Comparison of NSCs and ADSCs.
CharacteristicsNSCsADSCsReferences
SourcesEmbryonic NSCs (neural tube region); Adult NSCs (hippocampus, SVZ); ESCs; iPSCsAdipose tissue (subcutaneous fat)[17,49]
Morphological CharacteristicsForm neurospheres in specific culture medium; Spindle-shaped or multi-protrusion morphologyFibroblast-like, spindle-shaped morphology[30,57]
Differentiation PotentialNeurons; Astrocytes; OligodendrocytesAdipocytes; Osteoblasts; Chondrocytes; Hepatic lineage; Neural cells[34,58]
Surface MarkersNestin; Sox2; CD133; Musashi-1CD9, CD10, CD13, CD29, CD73, CD90, CD105, CD271; Do not express HSCs markers (CD31, CD45, CD11B)[41,59,60]
Neurotrophic factorsNGF, BDNF, GDNF, IGF-1; TGF-β; IGF1VEGF, EGF, HGF, IGF1, PGDF, FGF, TGF-β, BDNF, GDNF, NGF[52,61,62,63,64]
Proliferation CapacitySelf-renewal through symmetric and asymmetric divisionSelf-renewal and long-term proliferation capacity in vitro[18,56]
ImmunogenicityAllogeneic transplantation may trigger immune responsesSuitable for autologous transplantation[65,66]
Special effect-Secreting cytokines and exosomes;
EVs
[54,55,56,67,68]

3. Research Methods of ADSCs-to-iNSCs Induction Process

ADSCs have emerged as a promising source for iNSCs generation owing to their accessibility, multipotency, and lower immunogenicity [49,53]. Various induction methods were established for differentiation of ADSCs into iNSCs with specific advantages and limitations associated with each [11]. The most widely used induction strategies are described herein, such as chemical factors, growth factors, gene editing, 3D culture, co-culture, and combination approaches (Figure 2 and Table 2).

3.1. Chemical Induction

Chemical induction is achieved by using certain chemicals that stimulate signaling pathways to differentiate ADSCs into neural cells. Retinoic acid (RA), β-mercaptoethanol (BME), sertraline, valproic acid (VPA), butylated hydroxyanisole, forskolin, and L-carnitine (LC), etc., are some of the commonly used chemical inducers.
Retinoic acid (RA): RA, a metabolite of vitamin A, is a ligand for nuclear RA receptors (RARs) and an essential factor in neural development. It induces neural progenitors gene expression like Sox1 and Sox2, and suppresses mesodermal differentiation [69,70].
β-mercaptoethanol (BME): BME is a reducing agent that maintains the intracellular redox balance, supporting cell survival and differentiation [71]. BME pre-treatment and subsequent neural induction medium (NIM) have been shown to induce ADSCs into neural progenitor cells [72,73].
Valproic acid (VPA): VPA, an inhibitor of histone deacetylase, induces mature neuronal differentiation of ADSCs through the regulation of calcium signaling and nitric oxide pathways [74,75]. Furthermore, VPA improves the efficiency and specificity of induction by synergistically interacting with other chemical inducers [76].
L-carnitine (LC): LC, a derivative of amino acid [77], promotes neurogenic differentiation through the activation of Wnt/β-catenin and the protein kinase A (PKA) pathway [78].
Other chemical inducers: Sertraline is a synthetic chemical of the selective serotonin reuptake inhibitors (SSRIs) drug class and is used for the treatment of depression, anxiety, obsessive-compulsive disorder, and other psychosocial disorders [79]. It was found that sertraline promotes ASDCs’ proliferation and differentiation, whereas it inhibits the gliogenesis of ADSCs [80].
Chemical induction offers a cost-effective and straightforward approach to ASCDs’ differentiation into iNSCs. These chemicals activate the critical signaling pathways involved in neural development, making them accessible and easy-to-use reagents for researchers. The approach is, nonetheless, beset by drawbacks that include low specificity because the chemical may have off-target effects and needs careful optimization to achieve reproducible and efficient differentiation.

3.2. Growth Factors

Growth factors are essential for the activation of the in vivo microenvironment and to induce neural differentiation. The most significant growth factors are B27, basic fibroblast growth factor (bFGF), brain-derived neurotrophic factor (BDNF), epidermal growth factor (EGF) and human platelet lysate (HPL) [81,82].
The EGF and bFGF are commonly utilized in combination to induce neurosphere formation and maintain the self-renewal of neural progenitor cells [83]. In 2004, non-human primate adipose tissue stromal cells were successfully induced to develop into neurospheres in a B27, bFGF, BDNF and EGF-supplemented culture medium [84]. From 2014, Feng et al. set up a three-step induction protocol to differentiate very pure NSCs from human ADSCs through an activation of SOX1 by a conditioned medium with EGF and bFGF [81]. Later, ADSCs were induced to develop neurospheres with NSC-like properties through a neurobasal medium containing EGF and bFGF-2, and B27 [85,86,87,88]. More recently, homogenous cell populations of proliferating ADSCs cells have been induced to differentiate into iNSCs in culture media supplemented with EGF, bFGF, N2, and B27 [89]. ADSCs are differentiated to cells with a Schwann cell phenotype when cells are exposed to a combination of glial growth factors (GGF-2, bFGF, and PDGF) [90]. BDNF supports neuronal differentiation and survival through the activation of TrkB receptors [91]. Apart from these general growth factors, certain other factors, such as HPL and ghrelin, have also been utilized for differentiating ADSCs. Since HPL contains neurotrophic factors such as NTF3, BDGF, GDNF, and NGF, it is a potent inducer of neural differentiation [92].
While growth factors provide a very specific and effective means of inducing neural differentiation, their high cost and variability in stability, along with the need for stringent control over their concentration and timing, pose significant limitations to their large-scale application.

3.3. Gene Editing Technology

Gene editing techniques, for example, lentivirus or retrovirus transduction, enable targeted overexpression of neural-specific genes like Sox2, OCT4, and KLF4 in ADSCs, which trigger them to differentiate into iNSCs. These processes are highly specific and long-lasting due to stable genetic modification, making them efficient tools to yield functional neural cells. In addition, forced expression of Sox2 has been reported to directly induce the NSC phenotype in ADSCs [93], while multi-gene editing including OCT4, KLF4, Sox2, and c-MYC can reprogram ADSCs into iPSCs for subsequent neural differentiation [94]. However, gene editing is faced with challenges such as ethical concerns, technical complexity, and the risk of off-target effects or tumorigenicity, which need to be addressed carefully to ensure it offers safe and effective clinical translation.

3.4. Three-Dimensional (3D) Culture System

3D culture systems such as fibrin matrix and hydrogel scaffolds present a physiologically relevant environment mimicking the biochemical and mechanical properties of the central nervous system [95]. These systems enhance ADSC differentiation into iNSCs by promoting cell–cell interactions, neurosphere formation, and the expression of neural markers. For example, fibrin-based matrices induce stable ADSC differentiation into neural progenitor cells, while PEG-based hydrogels can induce spontaneous neural differentiation and enhance cell proliferation [96]. While 3D culture systems represent a more in vivo-like environment for neural differentiation, their complicated setup, scaffold characteristic variability, and requirement for precise optimization of mechanical and biochemical cues are challenges to standardization and scaling up in clinical applications.

3.5. Co-Culture Induction

Induction of co-culture allows ADSCs to differentiate into iNSCs through the utilization of direct or indirect interaction with other cell types, i.e., NSCs or olfactory ensheathing cells (OECs) [97]. Direct co-culture induces neural differentiation via physical cell–cell contact, whereas indirect co-culture uses soluble factors secreted from supporting cells to establish a neural-inductive microenvironment. For instance, co-culture of NSCs with ADSCs was found to improve neural marker expression and functional recovery [16], and OEC-conditioned medium is able to guide neural differentiation in the absence of direct contact [98]. While co-culture systems better mimic natural developmental signaling, they are limited by the availability of such specific cell types and are also problematic for standardization, thus rendered less feasible in large-scale applications [99].

3.6. Combined Induction

Combination methods integrate two or more induction approaches, including chemical reagents, growth factors, and gene editing, to differentiate ADSCs into iNSCs in an enhanced and targeted way. With combination strategies such as bFGF and EGF combined with chemicals or overexpression of neural-specific genes in a 3D culture environment, they realize the elevated expression of neural markers and functional characteristics via synergistic effects [81,100,101]. For instance, studies have indicated that the application of small molecules (e.g., SB431541 and noggin) together with growth factors (e.g., EGF and bFGF) improves the neural differentiation potential of ADSCs [102]. Although the combination approaches provide improved induction efficiency and functional integration, their greater complexity, higher cost, and requirement for precise optimization of multiple factors are barriers to clinical translation and scalability.
Table 2. Induction methods of ADSCs into iNSCs.
Table 2. Induction methods of ADSCs into iNSCs.
Induction MethodsKey Factors/TechniquesAdvantagesChallengesReferences
Chemical Factor RA; BME; Forskolin; Sertraline; VPA; VPA + butylated hydroxyanisole + insulin + hydrocortisone; LC; BMP4Cost-effective;
Easy to implement
Limited specificity;
Potential off-target effects
[50,70,72,73,74,75]
Growth Factor BDNF; GDNF; EGF; bFGF; NGF; TGF-β; N2; B27; Ghrelin; FGF2High specificity and efficacyHigh cost and potential instability of growth factors[81,85,86,87,88]
Gene Editing Sox2; CGRP; OCT4; KLF4; SOX2; and c-MYCHigh precision;
Long-lasting effects
Ethical concerns, off-target effects;
Technical complexity
[89,94,103]
3D Culture SystemFibrin matrix microenvironment; Hydrogel scaffold; PEG-Based 3D MatrixBetter mimics in vivo conditionsComplex setup;
Potential variability
[95,96,104]
Co-Culture InductionDirect contact co-culture; No-contact co-culture with ESCs; Chitosan co-culture SystemsUtilizes natural signaling mechanismsRequires access to other cells[16,99,105]
Combined InductionMelatonin + CM; Indomethacin + Insulin + IBMX + PBM; Sox1 Activation + CM; bFGF + forskolin; BDNF + RA; SB431542 + noggin + LDN193289 + EGF + bFGF; 3D hydrogels + B27 + C1; Insoluble fibrin supported adhesion matrix + growth factors;Maximizes induction efficiency/outcomesIncreased complexity and cost[81,100,101,102,106,107,108,109]
In summary, compounds, growth factors, gene editing, 3D culture, co-culture, and various combinations can induce ADSC differentiation into iNSCs (Figure 2 and Table 2). In addition, there are still some problems in validating the method of inducing ADSC differentiation. There are currently few in vivo studies on the differentiation of ADSC into iNSCs, and most studies only focus on the expression of surface markers, so the stemness and differentiation functions after induction have not been fully verified. Hence, the best combination of factors remains to be determined.

4. Molecular Signal Pathways of ADSCs-to-iNSCs Induction Process

The differentiation of ADSCs into iNSCs is governed by a complex molecular signaling network [110]. Chapter 3 outlines various approaches to induce stem cell differentiation, while this chapter delves into the signaling pathways that mediate these effects. Induction methods trigger specific signaling pathways that guide stem cells to their target fate, highlighting the interaction between external cues and cellular responses. Notch, Wnt/β-catenin, and Akt/mTOR pathways, among many others, control the process of reprogramming by influencing gene expression and cellular activities [111,112]. The following is an overview of the major pathways in ADSC-to-iNSC differentiation and their roles in neural development (Figure 3 and Table 3).

4.1. Notch Signaling Pathway

The Notch pathway is needed for maintaining the undifferentiated phenotype of iNSCs through the inhibition of premature neuronal differentiation [109]. Activation occurs when Notch receptors bind to ligands from neighboring cells, triggering proteolytic cleavage and release of the Notch intracellular domain (NICD). The NICD translocates to the nucleus, influencing gene expression by interacting with transcription factors [113]. In human ADSCs, notch signaling along with fibrin-based niche elements control the fate of neural progenitor cells [113,114]. Notch signaling inhibition was found to facilitate neuronal differentiation, thereby indicating its function as a gatekeeper of neural commitment [115].

4.2. Wnt/β-Catenin Signaling Pathway

Wnt signaling is crucial for stem cell self-renewal and differentiation. In the canonical pathway, Wnt proteins bind to Frizzled receptors, stabilizing β-catenin, which then enters the nucleus to modulate gene expression [116]. The Wnt/β-catenin pathway plays context-dependent roles in ADSCs differentiation. In rat ADSCs, ghrelin-activated Wnt/β-catenin strongly induces neurogenic differentiation [92]. The pathway also works together with fibrin-based matrices to cause neural progenitor cell proliferation and differentiation [109,117]. In addition, Wnt/β-catenin signaling also synergizes with other pathways such as PKA, to drive neural differentiation [118,119].

4.3. Akt/mTOR Signaling Pathway

The Akt/mTOR pathway regulates cell survival, growth, and differentiation [120]. Stimulating this pathway in ADSCs with ghrelin activates biphasic regulation of Wnt/β-catenin and mTOR signaling, thereby inducing neural differentiation [78,92,94,109]. Its crosstalk with Wnt and Notch pathways underscores its integrative role in ADSC-to-iNSC reprogramming. The pharmacological regulation of Akt/mTOR has also been reported to enhance the effectiveness of ADSC-to-iNSC differentiation and therefore represents a therapeutic target opportunity [92].

4.4. Calcium Signaling and Redox Regulation

Calcium signaling and redox regulation are critical modulators of ADSC differentiation. VPA-treated ADSCs have functional N-type voltage-gated Ca2+ channels, which induce mature neuronal commitment [75]. In addition, the VPA-induced iNOS-NO-sGC axis is involved in neural differentiation induction [74]. These findings highlight the importance of calcium and redox signaling in ADSC reprogramming.

4.5. Multi-Pathway Crosstalk

ADSCs reprogramming into iNSCs entails the hierarchical coordination of multiple signaling pathways. LC, for instance, induces neurogenesis via the simultaneous activation of PKA and Wnt/β-catenin [78], whereas ghrelin possesses dual regulatory roles via Wnt/β-catenin and Akt/mTOR [92]. Such interactions highlight the intricacy of ADSC reprogramming and system-level appreciation of signaling networks.
These pathways do not operate in isolation; they form a dynamic network that integrates external signals and intrinsic cellular states. The interplay between Notch, Wnt/β-catenin, and Akt/mTOR pathways exemplifies the complexity of molecular mechanisms driving ADSC-to-iNSC differentiation. A systematic understanding of these interactions provides insights into potential therapeutic strategies for enhancing neural differentiation. Each pathway plays a critical role in regulating stem cell fate, with the Notch pathway maintaining the undifferentiated state, while Wnt/β-catenin and Akt/mTOR pathways facilitate neurogenic differentiation through various stimuli. Additionally, the interactions among these pathways emphasize the intricate crosstalk involved in ADSC reprogramming, enhancing our knowledge of the neural potential of ADSCs and opening up avenues for improving ADSC-derived iNSC differentiation.
Table 3. Molecular mechanisms of ADSCs-to-iNSCs induction process. hADSCs: human ADSCs; rADSCs: rat ADSCs.
Table 3. Molecular mechanisms of ADSCs-to-iNSCs induction process. hADSCs: human ADSCs; rADSCs: rat ADSCs.
Signaling PathwaySourceInduction MethodsDescriptionReferences
Notch hADSCsBiomimetic nicheMaintains the undifferentiated state of iNSCs[107]
Wnt/β-cateninhADSCsBiomimetic nicheInduces cell proliferation[109]
rADSCsGhrelinPromotes neural differentiation[92]
rADSCsLCPromotes neural differentiation[78]
rADSCsCGRP gene-editingPromotes neural differentiation[94]
Calcium (Ca2+) and ROS rADSCsVPAPromotes neural differentiation[75]
iNOS-NO-sGCrADSCsVPAPromotes neural differentiation [74]
Akt/mTORrADSCsGhrelinPromotes neural differentiation[92]
PKArADSCsLCPromotes neural differentiation[78]

5. Application Prospects

To date, ADSCs and their induced iNSCs have shown great potential in the therapy of PD and other neurological disorders [121,122,123]. Their abundance, multipotency, and low immunogenicity render them an effective alternative to conventional stem cell sources. Here, the therapeutic prospectives of ADSCs and iNSCs for PD, their utility in drug and neurotoxicity assessment, and tissue engineering, as well as clinical translation challenges, will be elaborated (Table 4).

5.1. Therapeutic Potential in Parkinson’s Disease

ADSCs exhibited neuroprotection in PD preclinical models, such as the protection of dopaminergic neurons and inhibition of neuroinflammation [122,124]; especially, ADSC-EVs could inhibit the activation of microglia and protect neurons from apoptosis [125]. Also, ADSCs are alive, migrate, and become functionally integrated into damaged neural tissues upon transplantation, where they differentiate into tyrosine hydroxylase (TH)-positive dopaminergic neurons and ameliorate motor deficits [126,127]. In comparison to NSCs, ADSCs also have the advantages of being simply isolated from autologous adipose tissue, having low immunogenicity, and not requiring invasive brain surgery [128]. While NSCs have more robust neural differentiation capability, their use is hampered by ethical issues, risks of tumorigenicity, and poor survival of grafts [129].
In animal models, ADSCs have been induced to differentiate into dopaminergic neurons and restore motor dysfunction [123,130]. For example, ADSCs induced in a neurogenic differentiation medium and grafted into 6-hydroxydopamine (6-OHDA)-lesioned rats survived as dopaminergic neurons and restored the motor function of the animal [131]. Similarly, grafting of LMX1A-overexpressing ADSCs with adenoviral delivery of neurturin (NTN) and TH (Ad-NTN-TH) enhanced dopaminergic gene expression and conferred superior neuroprotection in MPTP-lesioned hemiparkinsonian rhesus monkeys [132]. These results demonstrate the potential of ADSCs as a treatment candidate for PD.

5.2. Drug and Neurotoxicity Assessment

Apart from direct cell therapy, ADSCs offer significant therapeutic potential for enhancing the efficacy of certain drugs. ADSCs loaded with palm oil ester-coated magnetic nanoparticles represent a promising strategy for targeted drug delivery to solid tumors, showing that ADSCs maintain high viability and motility while effectively carrying anti-tumor drugs [133]. Furthermore, ADSC-derived iNSCs also offer an invaluable tool for drug and neurotoxicity testing. The cells allow for in vitro screening of neuroprotective or regenerative medications, which diminishes the need for animal studies and avoids ethical restrictions [134]. For instance, iNSCs have been employed to model PD in vitro, which has enabled researchers to investigate disease mechanisms and screen for putative therapies in a controlled environment [135].

5.3. Neural Tissue Engineering

ADSC-derived iNSCs are also being investigated as precursor cells for neural tissue engineering. When used in combination with biomaterials like 3D-printed scaffolds, iNSCs facilitate nerve regeneration and increase axonal regrowth in injured tissues [95,100,136]. These strategies can reconstitute neural circuits and restore function in patients with PD and other neurological disorders.
Although ADSC-based therapies show promise, several issues must be addressed before they can be successfully implemented in clinical settings. These challenges include optimizing differentiation conditions to promote functional incorporation, resolving safety concerns related to tumorigenicity, and scaling up production for broader use. Additionally, long-term efficacy studies are essential to evaluate the effectiveness and safety of ADSC-derived iNSCs in human patients. In the next chapter, we will explore specific solutions to these challenges and outline future research directions.
Table 4. Therapeutic applications of ADSCs and iNSCs in neurological disorders.
Table 4. Therapeutic applications of ADSCs and iNSCs in neurological disorders.
ApplicationKey FindingsReferences
Dopaminergic neuron replacementADSCs differentiate into TH-positive neurons and improve motor deficits in PD models[126,127]
NeuroprotectionADSCs suppress neuroinflammation and preserve dopaminergic neurons[123,125,137]
Drug assessmentADSCs and iNSCs provide a platform for identifying neuroprotective compounds and drug delivery[57,133]
Neural tissue engineeringiNSCs combined with biomaterials promote nerve regeneration and axonal regrowth[136]

6. Research Challenges and Future Prospects

Although significant progress has been made in developing ADSC-induced iNSCs for PD treatment, some critical challenges have to be addressed before clinical implementation.

6.1. Technical Bottlenecks

The current induction protocols (Table 2) are highly heterogeneous, with differentiation efficiencies varying based on technique and donor variables. While chemical induction approaches using compounds like VPA and RA are relatively inexpensive and straightforward, they produce heterogeneous populations with partial neural commitment. Growth factor-based strategies have enhanced specificity but need precise optimization of time and dose to prevent overstimulation. Even more sophisticated techniques, including 3D cell culture and gene manipulation, more closely replicate physiology at the expense of increased complexity of standardization and scale.
Single-cell sequencing (scRNA-seq) studies have uncovered inherent ADSC heterogeneity as a significant limiting factor, with only 20–30% of cells attaining full neural reprogramming [103,138]. This heterogeneity is highly donor age- and harvest site-dependent [139,140,141,142]. Recent research has revealed the pseudo-temporal dynamic evolution characteristics of ADSCs during their differentiation into neuronal cells via scRNA-seq [143]. This approach facilitates the selection of specific neuronal cell types from ADSCs post-induction, effectively reducing heterogeneity.

6.2. Clinical Translation Challenges

The translation from preclinical studies to clinical applications has particular difficulties for ADSC-based therapies [144]. While phase II trials have moved forward with 1200 patients with the use of pluripotent stem cells [6], ADSC-derived neural precursors face unique difficulties. One of the key concerns is the stability of induced phenotypes because up to 40% of chemically derived “neural-like” cells can dedifferentiate back to their original state or exhibit stress-related artifacts [7,145]. Prolonged in vitro culture beyond passage 10 risks karyotypic abnormalities [146], and thus necessitate diligent monitoring of genomic integrity.
The stromal vascular fraction, which is obtained from adipose tissue, contains functionally heterogeneous subpopulations [147], and evidence suggests that the CD271+/CD146+ subsets have strong neural differentiation potential [139,148,149,150]. Such biological complexity highlights the need to standardize isolation and characterization protocols. In addition, the degree to which cell replacement and paracrine mechanisms underlie beneficial effects is unclear, which makes it more difficult to further optimize the dose and choose it for delivery strategy.

6.3. Future Directions

Several key priorities emerge for advancing ADSC-based neural therapies for PD. First and foremost is the requirement of protocol optimization. This involves the development of GMP complaint induction protocols with AI-backed quality control. Functional validation assays to enhance consistency and reliability of cell production are also important. Second, mechanistic studies founded on single-cell multi-omics approaches are required to identify molecular signatures for predicting successful neural differentiation and integration that provide deeper insights into the biological processes of ADSC reprogramming. Third, long-term safety studies (e.g., NCT03308565) must be finished, followed by efficacy studies in well-characterized patient populations to determine clinical significance and therapeutic benefit [151].
New technologies have particular potential for surmounting existing limitations. CRISPR-based gene editing could enhance neural differentiation efficiency, and 3D bioprinting may improve graft survival and integration [152,153]. Functionalized biomaterial scaffolds (e.g., bioactive matrix-coated nerve conduits) with neurotrophic factors (e.g., pro-survival small molecules) may provide permissive microenvironments for transplanted cells [154,155].
Effective translation of ADSC-derived therapies will require academic–industry collaboration to develop standardized protocols, establish safety and efficacy, and scale up manufacturing processes. Through methodical resolution of current challenges, clinical translation appears attainable for certain neurological indications.

7. Conclusions

The induction of ADSCs into iNSCs is a promising neural regenerative therapeutic strategy with the additional benefit of an available autologous cell source and inherent neural regenerative capacity. Induction protocols are currently efficient through the specific modulation of essential signaling pathways, including Notch, Wnt/b-catenin, and Akt/mTOR. Preclinical investigations have also illustrated their ability to promote functional recovery in animal models. Nevertheless, significant hurdles remain, notably in the standardization of protocols, large-scale production, and stringent safety verification. New technique innovations like scRNA-seq, CRISPR-mediated engineering, 3D-bioprinted neural scaffolds, and AI-supported quality control systems are surmounting these hurdles and limitations, and accelerating translational advancement.
ADSC-iNSC-based therapies are expected to advance to clinical trials in the near future. Not only do these therapies show promise for the replacement of dopaminergic neurons, but also for delivering neuroprotection. ADSC-iNSCs thus represent one illustration of convergence between regenerative medicine and precision neurology as a therapeutic strategy for neurodegenerative disease.

Author Contributions

Conceptualization, K.-h.C. and K.K.L.Y.; Literature collection and preparation, J.P.; Writing—original draft preparation, J.P. and Z.Z.; Writing—review and editing, J.P., Z.Z., M.L., K.-h.C. and K.K.L.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Golden Meditech Centre for NeuroRegeneration Sciences (Donation No. 179206) and the Cluster Research Matching Scheme of Hong Kong Baptist University (CRMS/23-24/04).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article.

Acknowledgments

The authors sincerely thank Zhanpeng Feng, Yichao Ou, and Songtao Qi from the Department of Neurosurgery, Nanfang Hospital, Southern Medical University, for their invaluable expertise in critically reviewing and advising on the manuscript sections “Application Prospects” and “Research Challenges and Future Prospects.” Their insightful advice significantly strengthened the translational implications and methodological rigor of this work. The authors declare that they have not used AI-generated work in this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ADAlzheimer’s disease
ADSCsadipose-derived stem cells
ASCsadult stem cells
BDNFbrain-derived neurotrophic factor
BMEβ-mercaptoethanol
bFGFbasic fibroblastic growth factor
C1CultureOne
Ca2+calcium
CGRPcalcitonin gene-related peptide
CNScentral nervous system
CNTFciliary neurotrophic factor
CMconditioned medium
EGFepidermal growth factor
ESCsembryonic stem cells
EVsextracellular vesicles
FGF-2fibroblast growth factor 2
GDNFglial-derived neurotrophic factor
hADSCshuman ADSCs
HGFhepatic growth factor
HPLhuman platelet lysate
hPSCshuman pluripotent stem cells
IBMX3-Isobutyl-1-methylxanthine
iNSCsinduced neural stem cell-like cells
iPSCsinduced pluripotent stem cells
LCL-carnitine
mRNAmessenger RNA
MSCsmesenchymal stem cells
NGFnerve growth factor
NICDNotch intracellular domain
NSCsneural stem cells
NT-3neurotrophin-3
NTNneurturin
OCDobsessive-compulsive disorder
PBMphotobiomodulation
PDParkinson’s disease
PDGFplatelet-derived growth factor
PKAprotein kinase A
qPCRquantitative polymerase chain reaction
RAretinoic acid
RARsRA receptors
RNAribonucleic acid
SCIspinal cord injury
scRNA-seqsingle-cell RNA sequencing
SSRIsselective serotonin reuptake inhibitors
SVFstromal vascular fraction
SVZsubventricular zone
TGF-βtransforming growth factor beta
VEGFvascular endothelial growth factor
VPAValproic Acid
WntWingless-related integration site

References

  1. Wilson, D.M.; Cookson, M.R.; Van Den Bosch, L.; Zetterberg, H.; Holtzman, D.M.; Dewachter, I. Hallmarks of Neurodegenerative Diseases. Cell 2023, 186, 693–714. [Google Scholar] [CrossRef] [PubMed]
  2. Ramesh, S.; Arachchige, A.S.P.M. Depletion of Dopamine in Parkinson’s Disease and Relevant Therapeutic Options: A Review of the Literature. AIMS Neurosci. 2023, 10, 200–231. [Google Scholar] [CrossRef] [PubMed]
  3. Tolosa, E.; Garrido, A.; Scholz, S.W.; Poewe, W. Challenges in the Diagnosis of Parkinson’s Disease. Lancet Neurol. 2021, 20, 385–397. [Google Scholar] [CrossRef] [PubMed]
  4. Zhong, Y.; Liu, H.; Liu, G.; Zhao, L.; Dai, C.; Liang, Y.; Du, J.; Zhou, X.; Mo, L.; Tan, C.; et al. A Review on Pathology, Mechanism, and Therapy for Cerebellum and Tremor in Parkinson’s Disease. Npj Park. Dis. 2022, 8, 82. [Google Scholar] [CrossRef]
  5. Zheng, Y.; Zhou, J.; Wang, Y.; Fan, F.; Liu, S.; Wang, Y. Neural Stem/Progenitor Cell Transplantation in Parkinson’s Rodent Animals: A Meta-Analysis and Systematic Review. Stem Cells Transl. Med. 2022, 11, 383–393. [Google Scholar] [CrossRef]
  6. Kirkeby, A.; Nelander, J.; Hoban, D.B.; Rogelius, N.; Bjartmarz, H.; Novo Nordisk Cell Therapy R&D; Storm, P.; Fiorenzano, A.; Adler, A.F.; Vale, S.; et al. Preclinical Quality, Safety, and Efficacy of a Human Embryonic Stem Cell-Derived Product for the Treatment of Parkinson’s Disease, STEM-PD. Cell Stem Cell 2023, 30, 1299–1314.e9. [Google Scholar] [CrossRef]
  7. Cerneckis, J.; Cai, H.; Shi, Y. Induced Pluripotent Stem Cells (iPSCs): Molecular Mechanisms of Induction and Applications. Signal Transduct. Target. Ther. 2024, 9, 112. [Google Scholar] [CrossRef]
  8. Alió Del Barrio, J.L.; De la Mata, A.; De Miguel, M.P.; Arnalich-Montiel, F.; Nieto-Miguel, T.; El Zarif, M.; Cadenas-Martín, M.; López-Paniagua, M.; Galindo, S.; Calonge, M.; et al. Corneal Regeneration Using Adipose-Derived Mesenchymal Stem Cells. Cells 2022, 11, 2549. [Google Scholar] [CrossRef]
  9. Umezawa, A.; Fukuda, A.; Horikawa, R.; Uchida, H.; Enosawa, S.; Oishi, Y.; Nakamura, N.; Sasaki, K.; Yanagi, Y.; Shimizu, S.; et al. First-in-Human Clinical Study of an Embryonic Stem Cell Product for Urea Cycle Disorders. Stem Cell Res. Ther. 2025, 16, 120. [Google Scholar] [CrossRef]
  10. Soltani, A.; Moradi, M.; Nejad, A.R.; Moradi, S.; Javandoost, E.; Nazari, H.; Jafarian, A. Adipose-Derived Stem Cells: Potentials, Availability and Market Size in Regenerative Medicine. Curr. Stem Cell Res. Ther. 2023, 18, 347–379. [Google Scholar] [CrossRef]
  11. Qin, Y.; Ge, G.; Yang, P.; Wang, L.; Qiao, Y.; Pan, G.; Yang, H.; Bai, J.; Cui, W.; Geng, D. An Update on Adipose-Derived Stem Cells for Regenerative Medicine: Where Challenge Meets Opportunity. Adv. Sci. 2023, 10, 2207334. [Google Scholar] [CrossRef] [PubMed]
  12. Choudhery, M.S.; Badowski, M.; Muise, A.; Pierce, J.; Harris, D.T. Cryopreservation of Whole Adipose Tissue for Future Use in Regenerative Medicine. J. Surg. Res. 2014, 187, 24–35. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, J.; Liu, Y.; Chen, Y.; Yuan, L.; Liu, H.; Wang, J.; Liu, Q.; Zhang, Y. Adipose-Derived Stem Cells: Current Applications and Future Directions in the Regeneration of Multiple Tissues. Stem Cells Int. 2020, 2020, 8810813. [Google Scholar] [CrossRef] [PubMed]
  14. Yang, L.; Shen, X.-M.; Wang, Z.-F.; Li, K.; Wang, W. The Notch Signalling Pathway and miRNA Regulation Play Important Roles in the Differentiation of Schwann Cells from Adipose-Derived Stem Cells. Lab. Investig. 2022, 102, 320–328. [Google Scholar] [CrossRef]
  15. Pelegri, N.G.; Milthorpe, B.K.; Gorrie, C.A.; Santos, J. Neurogenic Marker Expression in Differentiating Human Adipose Derived Adult Mesenchymal Stem Cells. Stem Cell Investig. 2023, 10, 7. [Google Scholar] [CrossRef]
  16. Kaminska, A.; Radoszkiewicz, K.; Rybkowska, P.; Wedzinska, A.; Sarnowska, A. Interaction of Neural Stem Cells (NSCs) and Mesenchymal Stem Cells (MSCs) as a Promising Approach in Brain Study and Nerve Regeneration. Cells 2022, 11, 1464. [Google Scholar] [CrossRef]
  17. Mannino, G.; Russo, C.; Maugeri, G.; Musumeci, G.; Vicario, N.; Tibullo, D.; Giuffrida, R.; Parenti, R.; Lo Furno, D. Adult Stem Cell Niches for Tissue Homeostasis. J. Cell. Physiol. 2022, 237, 239–257. [Google Scholar] [CrossRef]
  18. Tang, X.; Deng, P.; Li, L.; He, Y.; Wang, J.; Hao, D.; Yang, H. Advances in Genetically Modified Neural Stem Cell Therapy for Central Nervous System Injury and Neurological Diseases. Stem Cell Res. Ther. 2024, 15, 482. [Google Scholar] [CrossRef]
  19. David-Bercholz, J.; Kuo, C.T.; Deneen, B. Astrocyte and Oligodendrocyte Responses from the Subventricular Zone After Injury. Front. Cell. Neurosci. 2021, 15, 797553. [Google Scholar] [CrossRef]
  20. Dermitzakis, I.; Manthou, M.E.; Meditskou, S.; Tremblay, M.-È.; Petratos, S.; Zoupi, L.; Boziki, M.; Kesidou, E.; Simeonidou, C.; Theotokis, P. Origin and Emergence of Microglia in the CNS—An Interesting (Hi)Story of an Eccentric Cell. Curr. Issues Mol. Biol. 2023, 45, 2609–2628. [Google Scholar] [CrossRef]
  21. Li, Q.; Zhang, S.; Zheng, Y.; Wen, H.; Han, X.; Zhang, M.; Guan, W. Differentiation Potential of Neural Stem Cells Derived from Fetal Sheep. Anim. Cells Syst. 2017, 21, 233–240. [Google Scholar] [CrossRef] [PubMed]
  22. Iltis, A.S.; Koster, G.; Reeves, E.; Matthews, K.R.W. Ethical, Legal, Regulatory, and Policy Issues Concerning Embryoids: A Systematic Review of the Literature. Stem Cell Res. Ther. 2023, 14, 209. [Google Scholar] [CrossRef] [PubMed]
  23. Konno, M.; Hamabe, A.; Hasegawa, S.; Ogawa, H.; Fukusumi, T.; Nishikawa, S.; Ohta, K.; Kano, Y.; Ozaki, M.; Noguchi, Y.; et al. Adipose-Derived Mesenchymal Stem Cells and Regenerative Medicine. Dev. Growth Differ. 2013, 55, 309–318. [Google Scholar] [CrossRef] [PubMed]
  24. Ouji-Sageshima, N.; Hiyama, A.; Kumamoto, M.; Kitabatake, M.; Hara, A.; Furukawa, R.; Hontsu, S.; Kawaguchi, T.; Sawabata, N.; Muro, S.; et al. Adipose-Derived Mesenchymal Stem Cells (ADSCs) Have Anti-Fibrotic Effects on Lung Fibroblasts from Idiopathic Pulmonary Fibrosis (IPF) Patients. Cells 2024, 13, 2050. [Google Scholar] [CrossRef]
  25. Farahmand, Y.; Nabiuni, M.; Vafaei Mastanabad, M.; Sheibani, M.; Mahmood, B.S.; Obayes, A.M.; Asadi, F.; Davallou, R. The Exo-microRNA (miRNA) Signaling Pathways in Pathogenesis and Treatment of Stroke Diseases: Emphasize on Mesenchymal Stem Cells (MSCs). Cell Biochem. Funct. 2024, 42, e3917. [Google Scholar] [CrossRef]
  26. Sharma, S.; Muthu, S.; Jeyaraman, M.; Ranjan, R.; Jha, S.K. Translational Products of Adipose Tissue-Derived Mesenchymal Stem Cells: Bench to Bedside Applications. World J. Stem Cells 2021, 13, 1360–1381. [Google Scholar] [CrossRef]
  27. Czerwiec, K.; Zawrzykraj, M.; Deptuła, M.; Skoniecka, A.; Tymińska, A.; Zieliński, J.; Kosiński, A.; Pikuła, M. Adipose-Derived Mesenchymal Stromal Cells in Basic Research and Clinical Applications. Int. J. Mol. Sci. 2023, 24, 3888. [Google Scholar] [CrossRef]
  28. Seo, Y.; Shin, T.-H.; Kim, H.-S. Current Strategies to Enhance Adipose Stem Cell Function: An Update. Int. J. Mol. Sci. 2019, 20, 3827. [Google Scholar] [CrossRef]
  29. Pan, H.; Bao, L.; Ji, M.; Lyu, Z.; Qi, N.; Wu, Y. A Human Embryonic Stem Cell-Derived Neural Stem Cell Senescence Model Triggered by Oxidative Stress. Curr. Stem Cell Res. Ther. 2025. [Google Scholar] [CrossRef]
  30. Ou, Y.; Che, M.; Peng, J.; Zhou, M.; Wu, G.; Gong, H.; Li, K.; Wang, X.; Niu, P.; Qi, S.; et al. An Efficient Method for the Isolation and Cultivation of Hypothalamic Neural Stem/Progenitor Cells from Mouse Embryos. Front. Neuroanat. 2022, 16, 711138. [Google Scholar] [CrossRef]
  31. Vishwakarma, S.K.; Bardia, A.; Tiwari, S.K.; Paspala, S.A.B.; Khan, A.A. Current Concept in Neural Regeneration Research: NSCs Isolation, Characterization and Transplantation in Various Neurodegenerative Diseases and Stroke: A Review. J. Adv. Res. 2014, 5, 277–294. [Google Scholar] [CrossRef] [PubMed]
  32. Hsu, L.-C.; Peng, B.-Y.; Chen, M.-S.; Thalib, B.; Ruslin, M.; Tung, T.D.X.; Chou, H.-H.; Ou, K.-L. The Potential of the Stem Cells Composite Hydrogel Wound Dressings for Promoting Wound Healing and Skin Regeneration: In Vitro and In Vivo Evaluation. J. Biomed. Mater. Res. B Appl. Biomater. 2019, 107, 278–285. [Google Scholar] [CrossRef] [PubMed]
  33. Chang, S.-Y.; Lee, J.H.; Oh, S.C.; Lee, M.Y.; Lim, N.K. Human Fibroblast Growth Factor-Treated Adipose-Derived Stem Cells Facilitate Wound Healing and Revascularization in Rats with Streptozotocin-Induced Diabetes Mellitus. Cells 2023, 12, 1146. [Google Scholar] [CrossRef] [PubMed]
  34. Naderi, N.; Combellack, E.J.; Griffin, M.; Sedaghati, T.; Javed, M.; Findlay, M.W.; Wallace, C.G.; Mosahebi, A.; Butler, P.E.; Seifalian, A.M.; et al. The Regenerative Role of Adipose-Derived Stem Cells (ADSC) in Plastic and Reconstructive Surgery. Int. Wound J. 2017, 14, 112–124. [Google Scholar] [CrossRef]
  35. Ababneh, N.A.; Al-Kurdi, B.; Jamali, F.; Awidi, A. A Comparative Study of the Capability of MSCs Isolated from Different Human Tissue Sources to Differentiate into Neuronal Stem Cells and Dopaminergic-like Cells. PeerJ 2022, 10, e13003. [Google Scholar] [CrossRef]
  36. Hemmati, H.D.; Nakano, I.; Lazareff, J.A.; Masterman-Smith, M.; Geschwind, D.H.; Bronner-Fraser, M.; Kornblum, H.I. Cancerous Stem Cells Can Arise from Pediatric Brain Tumors. Proc. Natl. Acad. Sci. USA 2003, 100, 15178–15183. [Google Scholar] [CrossRef]
  37. Baba, Y.; Onishi-Sakamoto, S.; Ide, K.; Nishifuji, K. Nestin Is a Marker of Unipotent Embryonic and Adult Progenitors Differentiating into an Epithelial Cell Lineage of the Hair Follicles. Sci. Rep. 2022, 12, 17820. [Google Scholar] [CrossRef]
  38. Hagey, D.W.; Bergsland, M.; Muhr, J. SOX2 Transcription Factor Binding and Function. Dev. Camb. Engl. 2022, 149, dev200547. [Google Scholar] [CrossRef]
  39. Chen, Y.; Chen, Y.; Li, Q.; Liu, H.; Han, J.; Zhang, H.; Cheng, L.; Lin, G. Short C-Terminal Musashi-1 Proteins Regulate Pluripotency States in Embryonic Stem Cells. Cell Rep. 2023, 42, 113308. [Google Scholar] [CrossRef]
  40. Pleskač, P.; Fargeas, C.A.; Veselska, R.; Corbeil, D.; Skoda, J. Emerging Roles of Prominin-1 (CD133) in the Dynamics of Plasma Membrane Architecture and Cell Signaling Pathways in Health and Disease. Cell. Mol. Biol. Lett. 2024, 29, 41. [Google Scholar] [CrossRef]
  41. Khazaei, S.; Keshavarz, G.; Bozorgi, A.; Nazari, H.; Khazaei, M. Adipose Tissue-Derived Stem Cells: A Comparative Review on Isolation, Culture, and Differentiation Methods. Cell Tissue Bank. 2022, 23, 1–16. [Google Scholar] [CrossRef] [PubMed]
  42. Su, C.; Mo, J.; Dong, S.; Liao, Z.; Zhang, B.; Zhu, P. Integrinβ-1 in Disorders and Cancers: Molecular Mechanisms and Therapeutic Targets. Cell Commun. Signal. CCS 2024, 22, 71. [Google Scholar] [CrossRef] [PubMed]
  43. Gu, Y.; Mu, Z.; Chen, Y.; Wu, C.; Shi, J.; Bai, N. Therapeutic Potential of ADSCs in Diabetic Wounds: A Proteomics-Based Approach. Front. Cell Dev. Biol. 2024, 12, 1468220. [Google Scholar] [CrossRef] [PubMed]
  44. Jeng, K.-S.; Sheen, I.-S.; Lin, S.-S.; Leu, C.-M.; Chang, C.-F. The Role of Endoglin in Hepatocellular Carcinoma. Int. J. Mol. Sci. 2021, 22, 3208. [Google Scholar] [CrossRef]
  45. Papadopoulos, K.S.; Piperi, C.; Korkolopoulou, P. Clinical Applications of Adipose-Derived Stem Cell (ADSC) Exosomes in Tissue Regeneration. Int. J. Mol. Sci. 2024, 25, 5916. [Google Scholar] [CrossRef]
  46. Llorente, V.; Velarde, P.; Desco, M.; Gómez-Gaviro, M.V. Current Understanding of the Neural Stem Cell Niches. Cells 2022, 11, 3002. [Google Scholar] [CrossRef]
  47. Matsubara, S.; Matsuda, T.; Nakashima, K. Regulation of Adult Mammalian Neural Stem Cells and Neurogenesis by Cell Extrinsic and Intrinsic Factors. Cells 2021, 10, 1145. [Google Scholar] [CrossRef]
  48. De Gioia, R.; Biella, F.; Citterio, G.; Rizzo, F.; Abati, E.; Nizzardo, M.; Bresolin, N.; Comi, G.P.; Corti, S. Neural Stem Cell Transplantation for Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 3103. [Google Scholar] [CrossRef]
  49. Cecerska-Heryć, E.; Pękała, M.; Serwin, N.; Gliźniewicz, M.; Grygorcewicz, B.; Michalczyk, A.; Heryć, R.; Budkowska, M.; Dołęgowska, B. The Use of Stem Cells as a Potential Treatment Method for Selected Neurodegenerative Diseases: Review. Cell. Mol. Neurobiol. 2023, 43, 2643–2673. [Google Scholar] [CrossRef]
  50. Setiawan, A.M.; Kamarudin, T.A.; Abd Ghafar, N. The Role of BMP4 in Adipose-Derived Stem Cell Differentiation: A Minireview. Front. Cell Dev. Biol. 2022, 10, 1045103. [Google Scholar] [CrossRef]
  51. Jankowski, M.; Stefańska, K.; Suchodolski, M.; Dompe, C.; Wąsiatycz, G.; Kempisty, B.; Nowicki, M.; Roszak, M. Differential Regulation of Apoptosis-Related Genes during Long-Term Culture and Differentiation of Canine Adipose-Derived Stem Cells —A Functional Bioinformatical Analysis. Front. Genet. 2024, 15, 1515778. [Google Scholar] [CrossRef] [PubMed]
  52. Issa, S.; Fayoud, H.; Shaimardanova, A.; Sufianov, A.; Sufianova, G.; Solovyeva, V.; Rizvanov, A. Growth Factors and Their Application in the Therapy of Hereditary Neurodegenerative Diseases. Biomedicines 2024, 12, 1906. [Google Scholar] [CrossRef] [PubMed]
  53. Surico, P.L.; Scarabosio, A.; Miotti, G.; Grando, M.; Salati, C.; Parodi, P.C.; Spadea, L.; Zeppieri, M. Unlocking the Versatile Potential: Adipose-Derived Mesenchymal Stem Cells in Ocular Surface Reconstruction and Oculoplastics. World J. Stem Cells 2024, 16, 89–101. [Google Scholar] [CrossRef]
  54. Liu, Y.-Y.; Li, Y.; Wang, L.; Zhao, Y.; Yuan, R.; Yang, M.-M.; Chen, Y.; Zhang, H.; Zhou, F.-H.; Qian, Z.-R.; et al. Mesenchymal Stem Cell-Derived Exosomes Regulate Microglia Phenotypes: A Promising Treatment for Acute Central Nervous System Injury. Neural Regen. Res. 2023, 18, 1657–1665. [Google Scholar] [CrossRef] [PubMed]
  55. Zhou, B.; Chen, Q.; Zhang, Q.; Tian, W.; Chen, T.; Liu, Z. Therapeutic Potential of Adipose-Derived Stem Cell Extracellular Vesicles: From Inflammation Regulation to Tissue Repair. Stem Cell Res. Ther. 2024, 15, 249. [Google Scholar] [CrossRef]
  56. Ciervo, Y.; Gatto, N.; Allen, C.; Grierson, A.; Ferraiuolo, L.; Mead, R.J.; Shaw, P.J. Adipose-Derived Stem Cells Protect Motor Neurons and Reduce Glial Activation in Both In Vitro and In Vivo Models of ALS. Mol. Ther. Methods Clin. Dev. 2021, 21, 413–433. [Google Scholar] [CrossRef]
  57. Corrêa, N.C.R.; Kuligovski, C.; Paschoal, A.C.C.; Abud, A.P.R.; Rebelatto, C.L.K.; Leite, L.M.B.; Senegaglia, A.C.; Dallagiovanna, B.; de Aguiar, A.M. Human Adipose-Derived Stem Cells (ADSC) and Human Periodontal Ligament Stem Cells (PDLSC) as Cellular Substrates of a Toxicity Prediction Assay. Regul. Toxicol. Pharmacol. 2018, 92, 75–82. [Google Scholar] [CrossRef]
  58. Gao, Y.; Liu, H.; Zheng, Q.; Yang, L.; Cao, G.; Yuan, J.; Hu, S.; Li, Z. Modulating Efficient Differentiation of Neural Stem Cells into Neurons by Using Plasmonic Nanoparticles and the NIR II Irradiation to Boost Therapy of Parkinson’s Disease. Nano Today 2024, 57, 102392. [Google Scholar] [CrossRef]
  59. Andreotti, J.P.; Silva, W.N.; Costa, A.C.; Picoli, C.C.; Bitencourt, F.C.O.; Coimbra-Campos, L.M.C.; Resende, R.R.; Magno, L.A.V.; Romano-Silva, M.A.; Mintz, A.; et al. Neural Stem Cell Niche Heterogeneity. Semin. Cell Dev. Biol. 2019, 95, 42–53. [Google Scholar] [CrossRef]
  60. Salehi, H.; Amirpour, N.; Niapour, A.; Razavi, S. An Overview of Neural Differentiation Potential of Human Adipose Derived Stem Cells. Stem Cell Rev. Rep. 2016, 12, 26–41. [Google Scholar] [CrossRef]
  61. Rhode, S.C.; Beier, J.P.; Ruhl, T. Adipose Tissue Stem Cells in Peripheral Nerve Regeneration-In Vitro and In Vivo. J. Neurosci. Res. 2021, 99, 545–560. [Google Scholar] [CrossRef] [PubMed]
  62. Marsh, S.E.; Blurton-Jones, M. Neural Stem Cell Therapy for Neurodegenerative Disorders: The Role of Neurotrophic Support. Neurochem. Int. 2017, 106, 94–100. [Google Scholar] [CrossRef] [PubMed]
  63. Zimmermann, T.; Remmers, F.; Lutz, B.; Leschik, J. ESC-Derived BDNF-Overexpressing Neural Progenitors Differentially Promote Recovery in Huntington’s Disease Models by Enhanced Striatal Differentiation. Stem Cell Rep. 2016, 7, 693–706. [Google Scholar] [CrossRef] [PubMed]
  64. Xiong, L.-L.; Zou, Y.; Shi, Y.; Zhang, P.; Zhang, R.-P.; Dai, X.-J.; Liu, B.; Wang, T.-H. Tree Shrew Neural Stem Cell Transplantation Promotes Functional Recovery of Tree Shrews with a Hemi-Sectioned Spinal Cord Injury by Upregulating Nerve Growth Factor Expression. Int. J. Mol. Med. 2018, 41, 3267–3277. [Google Scholar] [CrossRef]
  65. Moon, S.; Hong, J.; Go, S.; Kim, B.-S. Immunomodulation for Tissue Repair and Regeneration. Tissue Eng. Regen. Med. 2023, 20, 389–409. [Google Scholar] [CrossRef]
  66. Liang, Z.; Zhou, H.; Tang, R.; Zhang, S.; Chen, X.; Pei, L. Autologous Transplantation of Adipose-Derived Stromal Cells Combined with Sevoflurane Ameliorates Acute Lung Injury Induced by Cecal Ligation and Puncture in Rats. Sci. Rep. 2020, 10, 13760. [Google Scholar] [CrossRef]
  67. Mukhamedshina, Y.O.; Gracheva, O.A.; Mukhutdinova, D.M.; Chelyshev, Y.A.; Rizvanov, A.A. Mesenchymal Stem Cells and the Neuronal Microenvironment in the Area of Spinal Cord Injury. Neural Regen. Res. 2019, 14, 227–237. [Google Scholar] [CrossRef]
  68. Shariati Najafabadi, S.; Amirpour, N.; Amini, S.; Zare, N.; Kazemi, M.; Salehi, H. Human Adipose Derived Stem Cell Exosomes Enhance the Neural Differentiation of PC12 Cells. Mol. Biol. Rep. 2021, 48, 5033–5043. [Google Scholar] [CrossRef]
  69. Ghyselinck, N.B.; Duester, G. Retinoic Acid Signaling Pathways. Dev. Camb. Engl. 2019, 146, dev167502. [Google Scholar] [CrossRef]
  70. Brown, G. Retinoic Acid Receptor Regulation of Decision-Making for Cell Differentiation. Front. Cell Dev. Biol. 2023, 11, 1182204. [Google Scholar] [CrossRef]
  71. Liu, J.; Wang, H.; Ren, W.; Zhou, Y.; Ye, Z.; Tan, W.-S. β-Mercaptoethanol Promotes Osteogenesis of Human Mesenchymal Stem Cells via Sirt1-ERK Pathway. Cytotechnology 2020, 72, 695–706. [Google Scholar] [CrossRef] [PubMed]
  72. Radhakrishnan, S.; Trentz, O.A.; Reddy, M.S.; Rela, M.; Kandasamy, M.; Sellathamby, S. In Vitro Transdifferentiation of Human Adipose Tissue-Derived Stem Cells to Neural Lineage Cells—A Stage-Specific Incidence. Adipocyte 2019, 8, 164–177. [Google Scholar] [CrossRef] [PubMed]
  73. Bağcı, F.Ö.; Özgörgülü, A.; Çiçek, G.; Özen, E.U.; Duman, S.; Aktan, T.M.; Reisli, I.; Bagci, F.O.; Özgörgülü, A.; Çiçek, G.; et al. Evaluation of the Interaction Between Wharton’s Jelly-Derived Mesenchymal Stem Cells and β-Mercaptoethanol. Cureus 2025, 17, e83115. [Google Scholar] [CrossRef] [PubMed]
  74. Okubo, T.; Fujimoto, S.; Hayashi, D.; Suzuki, T.; Sakaue, M.; Miyazaki, Y.; Tanaka, K.; Usami, M.; Takizawa, T. Valproic Acid Promotes Mature Neuronal Differentiation of Adipose Tissue-Derived Stem Cells through iNOS-NO-sGC Signaling Pathway. Nitric Oxide Biol. Chem. 2019, 93, 1–5. [Google Scholar] [CrossRef]
  75. Satoh, A.; Fujimoto, S.; Irie, T.; Suzuki, T.; Miyazaki, Y.; Tanaka, K.; Usami, M.; Takizawa, T. Valproic Acid Promotes Differentiation of Adipose Tissue-Derived Stem Cells to Neuronal Cells Selectively Expressing Functional N-Type Voltage-Gated Ca2+ Channels. Biochem. Biophys. Res. Commun. 2022, 589, 55–62. [Google Scholar] [CrossRef]
  76. Liu, S.; Tian, H.; Niu, Y.; Yu, C.; Xie, L.; Jin, Z.; Niu, W.; Ren, J.; Fu, L.; Yao, Z. Combined Cell Grafting and VPA Administration Facilitates Neural Repair through Axonal Regeneration and Synaptogenesis in Traumatic Brain Injury. Acta Biochim. Biophys. Sin. 2022, 54, 1289–1300. [Google Scholar] [CrossRef]
  77. Guerreiro, G.; Deon, M.; Becker, G.S.; Dos Reis, B.G.; Wajner, M.; Vargas, C.R. Neuroprotective Effects of L-Carnitine towards Oxidative Stress and Inflammatory Processes: A Review of Its Importance as a Therapeutic Drug in Some Disorders. Metab. Brain Dis. 2025, 40, 127. [Google Scholar] [CrossRef]
  78. Fathi, E.; Farahzadi, R.; Charoudeh, H.N. L-Carnitine Contributes to Enhancement of Neurogenesis from Mesenchymal Stem Cells through Wnt/β-Catenin and PKA Pathway. Exp. Biol. Med. 2017, 242, 482–486. [Google Scholar] [CrossRef]
  79. Santos-Cruz, L.F.; Campos-Aguilar, M.; Castañeda-Partida, L.; Sigrist-Flores, S.C.; Heres-Pulido, M.E.; Dueñas-García, I.E.; Piedra-Ibarra, E.; Jiménez-Flores, R.; Ponciano-Gómez, A. Impact of Larval Sertraline Exposure on Alternative Splicing in Neural Tissue of Adult Drosophila Melanogaster. Int. J. Mol. Sci. 2025, 26, 563. [Google Scholar] [CrossRef]
  80. Razavi, S.; Jahromi, M.; Amirpour, N.; Khosravizadeh, Z. Effect of Sertraline on Proliferation and Neurogenic Differentiation of Human Adipose-Derived Stem Cells. Adv. Biomed. Res. 2014, 3, 97. [Google Scholar] [CrossRef]
  81. Feng, N.; Han, Q.; Li, J.; Wang, S.; Li, H.; Yao, X.; Zhao, R.C. Generation of Highly Purified Neural Stem Cells from Human Adipose-Derived Mesenchymal Stem Cells by Sox1 Activation. Stem Cells Dev. 2014, 23, 515–529. [Google Scholar] [CrossRef] [PubMed]
  82. Gupta, A.; Singh, S. Potential Role of Growth Factors Controlled Release in Achieving Enhanced Neuronal Trans-Differentiation from Mesenchymal Stem Cells for Neural Tissue Repair and Regeneration. Mol. Neurobiol. 2022, 59, 983–1001. [Google Scholar] [CrossRef] [PubMed]
  83. Liao, C.; Guan, Y.; Zheng, J.; Wang, X.; Wang, M.; Zhu, Z.; Peng, Q.; Wang, H.-H.; Li, M. Development of Synthetic Modulator Enabling Long-Term Propagation and Neurogenesis of Human Embryonic Stem Cell-Derived Neural Progenitor Cells. Biol. Res. 2023, 56, 59. [Google Scholar] [CrossRef]
  84. Kang, S.K.; Putnam, L.A.; Ylostalo, J.; Popescu, I.R.; Dufour, J.; Belousov, A.; Bunnell, B.A. Neurogenesis of Rhesus Adipose Stromal Cells. J. Cell Sci. 2004, 117 Pt 18, 4289–4299. [Google Scholar] [CrossRef] [PubMed]
  85. Zhang, Y.; Liu, N.; Tang, Y.; Yang, E.; Dong, S.; Huang, M.; Pan, C.; Zhang, Y.; Zhang, P.; Chen, H.; et al. Efficient Generation of Neural Stem Cell-like Cells from Rat Adipose Derived Stem Cells after Lentiviral Transduction with Green Fluorescent Protein. Mol. Neurobiol. 2014, 50, 647–654. [Google Scholar] [CrossRef]
  86. Darvishi, M.; Tiraihi, T.; Mesbah-Namin, S.A.; Delshad, A.; Taheri, T. Motor Neuron Transdifferentiation of Neural Stem Cell from Adipose-Derived Stem Cell Characterized by Differential Gene Expression. Cell. Mol. Neurobiol. 2017, 37, 275–289. [Google Scholar] [CrossRef]
  87. Yang, E.; Liu, N.; Tang, Y.; Hu, Y.; Zhang, P.; Pan, C.; Dong, S.; Zhang, Y.; Tang, Z. Generation of Neurospheres from Human Adipose-Derived Stem Cells. BioMed Res. Int. 2015, 2015, 743714. [Google Scholar] [CrossRef]
  88. Lim, J.-H.; Koh, S.; Thomas, R.; Breen, M.; Olby, N.J. Evaluation of Gene Expression and DNA Copy Number Profiles of Adipose Tissue-Derived Stromal Cells and Consecutive Neurosphere-like Cells Generated from Dogs with Naturally Occurring Spinal Cord Injury. Am. J. Vet. Res. 2017, 78, 371–380. [Google Scholar] [CrossRef]
  89. Kruminis-Kaszkiel, E.; Osowski, A.; Bejer-Oleńska, E.; Dziekoński, M.; Wojtkiewicz, J. Differentiation of Human Mesenchymal Stem Cells from Wharton’s Jelly Towards Neural Stem Cells Using a Feasible and Repeatable Protocol. Cells 2020, 9, 739. [Google Scholar] [CrossRef]
  90. Kingham, P.J.; Kalbermatten, D.F.; Mahay, D.; Armstrong, S.J.; Wiberg, M.; Terenghi, G. Adipose-Derived Stem Cells Differentiate into a Schwann Cell Phenotype and Promote Neurite Outgrowth In Vitro. Exp. Neurol. 2007, 207, 267–274. [Google Scholar] [CrossRef]
  91. Jin, W. Regulation of BDNF-TrkB Signaling and Potential Therapeutic Strategies for Parkinson’s Disease. J. Clin. Med. 2020, 9, 257. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, G.-B.; Pan, Y.-M.; Liu, Y.-S.; Hu, J.-H.; Zhang, X.-D.; Zhang, D.-W.; Wang, Y.; Feng, Y.-K.; Yu, J.-B.; Cheng, Y.-X. Ghrelin Promotes Neural Differentiation of Adipose Tissue-Derived Mesenchymal Stem Cell via AKT/mTOR and β-Catenin Signaling Pathways. Kaohsiung J. Med. Sci. 2020, 36, 405–416. [Google Scholar] [CrossRef] [PubMed]
  93. Qin, Y.; Zhou, C.; Wang, N.; Yang, H.; Gao, W.-Q. Conversion of Adipose Tissue-Derived Mesenchymal Stem Cells to Neural Stem Cell-Like Cells by a Single Transcription Factor, Sox2. Cell. Reprogramming 2015, 17, 221–226. [Google Scholar] [CrossRef] [PubMed]
  94. Yang, Q.; Du, X.; Fang, Z.; Xiong, W.; Li, G.; Liao, H.; Xiao, J.; Wang, G.; Li, F. Effect of Calcitonin Gene-Related Peptide on the Neurogenesis of Rat Adipose-Derived Stem Cells In Vitro. PLoS ONE 2014, 9, e86334. [Google Scholar] [CrossRef]
  95. Chandrababu, K.; Sreelatha, H.V.; Sudhadevi, T.; Anil, A.; Arumugam, S.; Krishnan, L.K. In Vivo Neural Tissue Engineering Using Adipose-Derived Mesenchymal Stem Cells and Fibrin Matrix. J. Spinal Cord. Med. 2023, 46, 262–276. [Google Scholar] [CrossRef]
  96. Gomila Pelegri, N.; Stanczak, A.M.; Bottomley, A.L.; Milthorpe, B.K.; Gorrie, C.A.; Padula, M.P.; Santos, J. Adipose-Derived Stem Cells Spontaneously Express Neural Markers When Grown in a PEG-Based 3D Matrix. Int. J. Mol. Sci. 2023, 24, 12139. [Google Scholar] [CrossRef]
  97. Liu, R.; Meng, X.; Yu, X.; Wang, G.; Dong, Z.; Zhou, Z.; Qi, M.; Yu, X.; Ji, T.; Wang, F. From 2D to 3D Co-Culture Systems: A Review of Co-Culture Models to Study the Neural Cells Interaction. Int. J. Mol. Sci. 2022, 23, 13116. [Google Scholar] [CrossRef]
  98. Lo Furno, D.; Mannino, G.; Pellitteri, R.; Zappalà, A.; Parenti, R.; Gili, E.; Vancheri, C.; Giuffrida, R. Conditioned Media from Glial Cells Promote a Neural-Like Connexin Expression in Human Adipose-Derived Mesenchymal Stem Cells. Front. Physiol. 2018, 9, 1742. [Google Scholar] [CrossRef]
  99. Bahmani, L.; Taha, M.F.; Javeri, A. Coculture with Embryonic Stem Cells Improves Neural Differentiation of Adipose Tissue-Derived Stem Cells. Neuroscience 2014, 272, 229–239. [Google Scholar] [CrossRef]
  100. Gomila Pelegri, N.; Stanczak, A.M.; Bottomley, A.L.; Cummins, M.L.; Milthorpe, B.K.; Gorrie, C.A.; Padula, M.P.; Santos, J. Neural Marker Expression in Adipose-Derived Stem Cells Grown in PEG-Based 3D Matrix Is Enhanced in the Presence of B27 and CultureOne Supplements. Int. J. Mol. Sci. 2023, 24, 16269. [Google Scholar] [CrossRef]
  101. Romano, I.R.; D’Angeli, F.; Gili, E.; Fruciano, M.; Lombardo, G.A.G.; Mannino, G.; Vicario, N.; Russo, C.; Parenti, R.; Vancheri, C.; et al. Melatonin Enhances Neural Differentiation of Adipose-Derived Mesenchymal Stem Cells. Int. J. Mol. Sci. 2024, 25, 4891. [Google Scholar] [CrossRef] [PubMed]
  102. Park, J.; Lee, N.; Lee, J.; Choe, E.K.; Kim, M.K.; Lee, J.; Byun, M.S.; Chon, M.-W.; Kim, S.W.; Lee, C.J.; et al. Small Molecule-Based Lineage Switch of Human Adipose-Derived Stem Cells into Neural Stem Cells and Functional GABAergic Neurons. Sci. Rep. 2017, 7, 10166. [Google Scholar] [CrossRef] [PubMed]
  103. Cairns, D.M.; Chwalek, K.; Moore, Y.E.; Kelley, M.R.; Abbott, R.D.; Moss, S.; Kaplan, D.L. Expandable and Rapidly Differentiating Human Induced Neural Stem Cell Lines for Multiple Tissue Engineering Applications. Stem Cell Rep. 2016, 7, 557–570. [Google Scholar] [CrossRef] [PubMed]
  104. Georgiou, M.; Golding, J.P.; Loughlin, A.J.; Kingham, P.J.; Phillips, J.B. Engineered Neural Tissue with Aligned, Differentiated Adipose-Derived Stem Cells Promotes Peripheral Nerve Regeneration across a Critical Sized Defect in Rat Sciatic Nerve. Biomaterials 2015, 37, 242–251. [Google Scholar] [CrossRef]
  105. Lo Furno, D.; Mannino, G.; Giuffrida, R.; Gili, E.; Vancheri, C.; Tarico, M.S.; Perrotta, R.E.; Pellitteri, R. Neural Differentiation of Human Adipose-Derived Mesenchymal Stem Cells Induced by Glial Cell Conditioned Media. J. Cell. Physiol. 2018, 233, 7091–7100. [Google Scholar] [CrossRef]
  106. Abrahamse, H.; Crous, A. Photobiomodulation Effects on Neuronal Transdifferentiation of Immortalized Adipose-Derived Mesenchymal Stem Cells. Lasers Med. Sci. 2024, 39, 257. [Google Scholar] [CrossRef]
  107. Jang, S.; Cho, H.-H.; Cho, Y.-B.; Park, J.-S.; Jeong, H.-S. Functional Neural Differentiation of Human Adipose Tissue-Derived Stem Cells Using bFGF and Forskolin. BMC Cell Biol. 2010, 11, 25. [Google Scholar] [CrossRef]
  108. Anghileri, E.; Marconi, S.; Pignatelli, A.; Cifelli, P.; Galié, M.; Sbarbati, A.; Krampera, M.; Belluzzi, O.; Bonetti, B. Neuronal Differentiation Potential of Human Adipose-Derived Mesenchymal Stem Cells. Stem Cells Dev. 2008, 17, 909–916. [Google Scholar] [CrossRef]
  109. Chandrababu, K.; Senan, M.; Krishnan, L.K. Exploitation of Fibrin-Based Signaling Niche for Deriving Progenitors from Human Adipose-Derived Mesenchymal Stem Cells towards Potential Neural Engineering Applications. Sci. Rep. 2020, 10, 7116. [Google Scholar] [CrossRef]
  110. Schäffler, A.; Büchler, C. Concise Review: Adipose Tissue-Derived Stromal Cells—Basic and Clinical Implications for Novel Cell-Based Therapies. Stem Cells 2007, 25, 818–827. [Google Scholar] [CrossRef]
  111. Yang, K.; Wang, X.; Zhang, H.; Wang, Z.; Nan, G.; Li, Y.; Zhang, F.; Mohammed, M.K.; Haydon, R.C.; Luu, H.H.; et al. The Evolving Roles of Canonical WNT Signaling in Stem Cells and Tumorigenesis: Implications in Targeted Cancer Therapies. Lab. Investig. J. Tech. Methods Pathol. 2016, 96, 116–136. [Google Scholar] [CrossRef] [PubMed]
  112. Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/β-Catenin Signalling: Function, Biological Mechanisms, and Therapeutic Opportunities. Signal Transduct. Target. Ther. 2022, 7, 3. [Google Scholar] [CrossRef] [PubMed]
  113. Zhou, B.; Lin, W.; Long, Y.; Yang, Y.; Zhang, H.; Wu, K.; Chu, Q. Notch Signaling Pathway: Architecture, Disease, and Therapeutics. Signal Transduct. Target. Ther. 2022, 7, 95. [Google Scholar] [CrossRef] [PubMed]
  114. Mall, M.; Kareta, M.S.; Chanda, S.; Ahlenius, H.; Perotti, N.; Zhou, B.; Grieder, S.D.; Ge, X.; Drake, S.; Euong Ang, C.; et al. Myt1l Safeguards Neuronal Identity by Actively Repressing Many Non-Neuronal Fates. Nature 2017, 544, 245–249. [Google Scholar] [CrossRef]
  115. Borghese, L.; Dolezalova, D.; Opitz, T.; Haupt, S.; Leinhaas, A.; Steinfarz, B.; Koch, P.; Edenhofer, F.; Hampl, A.; Brüstle, O. Inhibition of Notch Signaling in Human Embryonic Stem Cell–Derived Neural Stem Cells Delays G1/S Phase Transition and Accelerates Neuronal Differentiation In Vitro and In Vivo. Stem Cells 2010, 28, 955–964. [Google Scholar] [CrossRef]
  116. Leung, R.W.H.; Lee, T.K.W. Wnt/β-Catenin Signaling as a Driver of Stemness and Metabolic Reprogramming in Hepatocellular Carcinoma. Cancers 2022, 14, 5468. [Google Scholar] [CrossRef]
  117. Yu, M.; Qin, K.; Fan, J.; Zhao, G.; Zhao, P.; Zeng, W.; Chen, C.; Wang, A.; Wang, Y.; Zhong, J.; et al. The Evolving Roles of Wnt Signaling in Stem Cell Proliferation and Differentiation, the Development of Human Diseases, and Therapeutic Opportunities. Genes Dis. 2024, 11, 101026. [Google Scholar] [CrossRef]
  118. Kang, D.E.; Soriano, S.; Xia, X.; Eberhart, C.G.; Strooper, B.D.; Zheng, H.; Koo, E.H. Presenilin Couples the Paired Phosphorylation of β-Catenin Independent of Axin: Implications for β-Catenin Activation in Tumorigenesis. Cell 2002, 110, 751–762. [Google Scholar] [CrossRef]
  119. Austin, S.H.L.; Gabarró-Solanas, R.; Rigo, P.; Paun, O.; Harris, L.; Guillemot, F.; Urbán, N. Wnt/β-Catenin Signalling Is Dispensable for Adult Neural Stem Cell Homeostasis and Activation. Dev. Camb. Engl. 2021, 148, dev199629. [Google Scholar] [CrossRef]
  120. Glaviano, A.; Foo, A.S.C.; Lam, H.Y.; Yap, K.C.H.; Jacot, W.; Jones, R.H.; Eng, H.; Nair, M.G.; Makvandi, P.; Geoerger, B.; et al. PI3K/AKT/mTOR Signaling Transduction Pathway and Targeted Therapies in Cancer. Mol. Cancer 2023, 22, 138. [Google Scholar] [CrossRef]
  121. Liu, M.; Lu, Y.; Sun, F.; Li, Y.; Wu, J.; Zou, Q. The Nerve-Induced Adipose Stem Cells Promote Nerve Repair in Stress Urinary Incontinence by Regulating Schwann Cell Repair Phenotype Conversion Through Activation of the Notch Pathway. Mol. Neurobiol. 2025, 62, 7330–7344. [Google Scholar] [CrossRef] [PubMed]
  122. Li, K.; Li, X.; Shi, G.; Lei, X.; Huang, Y.; Bai, L.; Qin, C. Effectiveness and Mechanisms of Adipose-Derived Stem Cell Therapy in Animal Models of Parkinson’s Disease: A Systematic Review and Meta-Analysis. Transl. Neurodegener. 2021, 10, 14. [Google Scholar] [CrossRef] [PubMed]
  123. McCoy, M.K.; Martinez, T.N.; Ruhn, K.A.; Wrage, P.C.; Keefer, E.W.; Botterman, B.R.; Tansey, K.E.; Tansey, M.G. Autologous Transplants of Adipose-Derived Adult Stromal (ADAS) Cells Afford Dopaminergic Neuroprotection in a Model of Parkinson’s Disease. Exp. Neurol. 2008, 210, 14–29. [Google Scholar] [CrossRef] [PubMed]
  124. Tambe, P.; Undale, V.; Sanap, A.; Bhonde, R.; Mante, N. The Prospective Role of Mesenchymal Stem Cells in Parkinson’s Disease. Park. Relat. Disord. 2024, 127, 107087. [Google Scholar] [CrossRef]
  125. Feng, N.; Huang, X.; Jia, Y. Small Extracellular Vesicles from Adipose Derived Stem Cells Alleviate Microglia Activation and Improve Motor Deficit of Parkinson’s Disease via miR-100-5p/DTX3L/STAT1 Signaling Axis. Exp. Neurol. 2025, 389, 115250. [Google Scholar] [CrossRef]
  126. Faghih, H.; Javeri, A.; Amini, H.; Taha, M.F. Directed Differentiation of Human Adipose Tissue-Derived Stem Cells to Dopaminergic Neurons in Low-Serum and Serum-Free Conditions. Neurosci. Lett. 2019, 708, 134353. [Google Scholar] [CrossRef]
  127. Hamedi, H.; Ghorbanian, S.; Mirzaeian, L.; Abrari, K.; Mozdziak, P.; Ghorbanian, M.T. Intravenous Transplantation of Adipose-Derived Mesenchymal Stem Cells Promoted the Production of Dopaminergic Neurons and Improved Spatial Memory in A Rat Model of Parkinson’s Disease. Cell J. Yakhteh 2023, 25, 317–326. [Google Scholar] [CrossRef]
  128. Jankowski, M.; Dompe, C.; Sibiak, R.; Wąsiatycz, G.; Mozdziak, P.; Jaśkowski, J.M.; Antosik, P.; Kempisty, B.; Dyszkiewicz-Konwińska, M. In Vitro Cultures of Adipose-Derived Stem Cells: An Overview of Methods, Molecular Analyses, and Clinical Applications. Cells 2020, 9, 1783. [Google Scholar] [CrossRef]
  129. Zhang, M.; Liu, Y.; Shi, L.; Fang, L.; Xu, L.; Cao, Y. Neural Stemness Unifies Cell Tumorigenicity and Pluripotent Differentiation Potential. J. Biol. Chem. 2022, 298, 102106. [Google Scholar] [CrossRef]
  130. Chi, K.; Fu, R.-H.; Huang, Y.-C.; Chen, S.-Y.; Hsu, C.-J.; Lin, S.-Z.; Tu, C.-T.; Chang, L.-H.; Wu, P.-A.; Liu, S.-P. Adipose-Derived Stem Cells Stimulated with n-Butylidenephthalide Exhibit Therapeutic Effects in a Mouse Model of Parkinson’s Disease. Cell Transplant. 2018, 27, 456–470. [Google Scholar] [CrossRef]
  131. Takahashi, H.; Ishikawa, H.; Tanaka, A. Regenerative Medicine for Parkinson’s Disease Using Differentiated Nerve Cells Derived from Human Buccal Fat Pad Stem Cells. Hum. Cell 2017, 30, 60–71. [Google Scholar] [CrossRef] [PubMed]
  132. Zhou, Y.; Sun, M.; Li, H.; Yan, M.; He, Z.; Wang, W.; Wang, W.; Lu, S. Recovery of Behavioral Symptoms in Hemi-Parkinsonian Rhesus Monkeys through Combined Gene and Stem Cell Therapy. Cytotherapy 2013, 15, 467–480. [Google Scholar] [CrossRef] [PubMed]
  133. Herea, D.-D.; Labusca, L.; Radu, E.; Chiriac, H.; Grigoras, M.; Panzaru, O.D.; Lupu, N. Human Adipose-Derived Stem Cells Loaded with Drug-Coated Magnetic Nanoparticles for In-Vitro Tumor Cells Targeting. Mater. Sci. Eng. C 2019, 94, 666–676. [Google Scholar] [CrossRef] [PubMed]
  134. Juberg, D.R.; Fox, D.A.; Forcelli, P.A.; Kacew, S.; Lipscomb, J.C.; Saghir, S.A.; Sherwin, C.M.; Koenig, C.M.; Hays, S.M.; Kirman, C.R. A Perspective on In Vitro Developmental Neurotoxicity Test Assay Results: An Expert Panel Review. Regul. Toxicol. Pharmacol. 2023, 143, 105444. [Google Scholar] [CrossRef]
  135. Wu, H.; Fan, Y.; Zhang, M. Advanced Progress in the Role of Adipose-Derived Mesenchymal Stromal/Stem Cells in the Application of Central Nervous System Disorders. Pharmaceutics 2023, 15, 2637. [Google Scholar] [CrossRef]
  136. Nakajima, T.; Tada, K.; Nakada, M.; Matsuta, M.; Tsuchiya, H. Facilitatory Effects of Artificial Nerve Filled with Adipose-Derived Stem Cell Sheets on Peripheral Nerve Regeneration: An Experimental Study. J. Orthop. Sci. 2021, 26, 1113–1118. [Google Scholar] [CrossRef]
  137. Berg, J.; Roch, M.; Altschüler, J.; Winter, C.; Schwerk, A.; Kurtz, A.; Steiner, B. Human Adipose-Derived Mesenchymal Stem Cells Improve Motor Functions and Are Neuroprotective in the 6-Hydroxydopamine-Rat Model for Parkinson’s Disease When Cultured in Monolayer Cultures but Suppress Hippocampal Neurogenesis and Hippocampal Memory Function When Cultured in Spheroids. Stem Cell Rev. Rep. 2015, 11, 133–149. [Google Scholar] [CrossRef]
  138. Isaković, J.; Šerer, K.; Barišić, B.; Mitrečić, D. Mesenchymal Stem Cell Therapy for Neurological Disorders: The Light or the Dark Side of the Force? Front. Bioeng. Biotechnol. 2023, 11, 1139359. [Google Scholar] [CrossRef]
  139. Câmara, D.A.D.; Shibli, J.A.; Müller, E.A.; De-Sá-Junior, P.L.; Porcacchia, A.S.; Blay, A.; Lizier, N.F. Adipose Tissue-Derived Stem Cells: The Biologic Basis and Future Directions for Tissue Engineering. Materials 2020, 13, 3210. [Google Scholar] [CrossRef]
  140. Saffari, T.M.; Saffari, S.; Vyas, K.S.; Mardini, S.; Shin, A.Y. Role of Adipose Tissue Grafting and Adipose-Derived Stem Cells in Peripheral Nerve Surgery. Neural Regen. Res. 2022, 17, 2179–2184. [Google Scholar] [CrossRef]
  141. Dong, L.; Li, X.; Leng, W.; Guo, Z.; Cai, T.; Ji, X.; Xu, C.; Zhu, Z.; Lin, J. Adipose Stem Cells in Tissue Regeneration and Repair: From Bench to Bedside. Regen. Ther. 2023, 24, 547–560. [Google Scholar] [CrossRef] [PubMed]
  142. Farhana, S.; Kai, Y.C.; Kadir, R.; Sulaiman, W.A.W.; Nordin, N.A.; Nasir, N.A.M. The Fate of Adipose Tissue and Adipose-Derived Stem Cells in Allograft. Cell Tissue Res. 2023, 394, 269–292. [Google Scholar] [CrossRef] [PubMed]
  143. Yuan, X.; Li, W.; Liu, Q.; Ou, Y.; Li, J.; Yan, Q.; Zhang, P. Single-Cell RNA-Seq Reveals the Pseudo-Temporal Dynamic Evolution Characteristics of ADSCs to Neuronal Differentiation. Cell. Mol. Neurobiol. 2024, 45, 5. [Google Scholar] [CrossRef] [PubMed]
  144. Toyserkani, N.M.; Jørgensen, M.G.; Tabatabaeifar, S.; Jensen, C.H.; Sheikh, S.P.; Sørensen, J.A. Concise Review: A Safety Assessment of Adipose-Derived Cell Therapy in Clinical Trials: A Systematic Review of Reported Adverse Events. Stem Cells Transl. Med. 2017, 6, 1786–1794. [Google Scholar] [CrossRef]
  145. Merrell, A.J.; Stanger, B.Z. Adult Cell Plasticity In Vivo: Trans-Differentiation Is Back in Style. Nat. Rev. Mol. Cell Biol. 2016, 17, 413–425. [Google Scholar] [CrossRef]
  146. Li, J.; Wu, Z.; Zhao, L.; Liu, Y.; Su, Y.; Gong, X.; Liu, F.; Zhang, L. The Heterogeneity of Mesenchymal Stem Cells: An Important Issue to Be Addressed in Cell Therapy. Stem Cell Res. Ther. 2023, 14, 381. [Google Scholar] [CrossRef]
  147. Cremona, M.; Gallazzi, M.; Rusconi, G.; Mariotta, L.; Gola, M.; Soldati, G. State of the Art in the Standardization of Stromal Vascular Fraction Processing. Biomolecules 2025, 15, 199. [Google Scholar] [CrossRef]
  148. van Strien, M.E.; Sluijs, J.A.; Reynolds, B.A.; Steindler, D.A.; Aronica, E.; Hol, E.M. Isolation of Neural Progenitor Cells from the Human Adult Subventricular Zone Based on Expression of the Cell Surface Marker CD271. Stem Cells Transl. Med. 2014, 3, 470–480. [Google Scholar] [CrossRef]
  149. Fayazi, M.; Salehnia, M.; Ziaei, S. Differentiation of Human CD146-Positive Endometrial Stem Cells to Adipogenic-, Osteogenic-, Neural Progenitor-, and Glial-like Cells. In Vitro Cell. Dev. Biol. Anim. 2015, 51, 408–414. [Google Scholar] [CrossRef]
  150. Figiel-Dabrowska, A.; Radoszkiewicz, K.; Rybkowska, P.; Krzesniak, N.E.; Sulejczak, D.; Sarnowska, A. Neurogenic and Neuroprotective Potential of Stem/Stromal Cells Derived from Adipose Tissue. Cells 2021, 10, 1475. [Google Scholar] [CrossRef]
  151. Bydon, M. Phase I Clinical Trial of Autologous Adipose Derived Mesenchymal Stem Cells in the Treatment of Paralysis Due to Traumatic Spinal Cord Injury; Clinical Trial Registration NCT03308565; clinicaltrials.gov. 2022. Available online: https://clinicaltrials.gov/study/NCT03308565 (accessed on 31 March 2025).
  152. Ahmed, M.; Muffat, J.; Li, Y. Understanding Neural Development and Diseases Using CRISPR Screens in Human Pluripotent Stem Cell-Derived Cultures. Front. Cell Dev. Biol. 2023, 11, 1158373. [Google Scholar] [CrossRef] [PubMed]
  153. Marei, H.E. Stem Cell Therapy: A Revolutionary Cure or a Pandora’s Box. Stem Cell Res. Ther. 2025, 16, 255. [Google Scholar] [CrossRef] [PubMed]
  154. Magaz, A.; Faroni, A.; Gough, J.E.; Reid, A.J.; Li, X.; Blaker, J.J. Bioactive Silk-Based Nerve Guidance Conduits for Augmenting Peripheral Nerve Repair. Adv. Healthc. Mater. 2018, 7, e1800308. [Google Scholar] [CrossRef] [PubMed]
  155. Poongodi, R.; Chen, Y.-L.; Yang, T.-H.; Huang, Y.-H.; Yang, K.D.; Lin, H.-C.; Cheng, J.-K. Bio-Scaffolds as Cell or Exosome Carriers for Nerve Injury Repair. Int. J. Mol. Sci. 2021, 22, 13347. [Google Scholar] [CrossRef]
Figure 1. Morphological characteristics of NSCs (A) and ADSCs (B) from adult rats (scale bar: 50 µm).
Figure 1. Morphological characteristics of NSCs (A) and ADSCs (B) from adult rats (scale bar: 50 µm).
Ijms 26 06599 g001
Figure 2. Induction processes of ADSCs-to-iNSCs, including chemicals, growth factors, gene editing technology, three-dimensional culture system, co-culture and combined induction method; the detailed protocols are contained in Table 2.
Figure 2. Induction processes of ADSCs-to-iNSCs, including chemicals, growth factors, gene editing technology, three-dimensional culture system, co-culture and combined induction method; the detailed protocols are contained in Table 2.
Ijms 26 06599 g002
Figure 3. Molecular signal pathways of ADSCs-to-iNSCs induction process.
Figure 3. Molecular signal pathways of ADSCs-to-iNSCs induction process.
Ijms 26 06599 g003
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

Peng, J.; Zhang, Z.; Li, M.; Yung, K.K.L.; Cheung, K.-h. From Adipose to Action: Reprogramming Stem Cells for Functional Neural Progenitors for Neural Regenerative Therapy. Int. J. Mol. Sci. 2025, 26, 6599. https://doi.org/10.3390/ijms26146599

AMA Style

Peng J, Zhang Z, Li M, Yung KKL, Cheung K-h. From Adipose to Action: Reprogramming Stem Cells for Functional Neural Progenitors for Neural Regenerative Therapy. International Journal of Molecular Sciences. 2025; 26(14):6599. https://doi.org/10.3390/ijms26146599

Chicago/Turabian Style

Peng, Junjie, Zhu Zhang, Min Li, Ken Kin Lam Yung, and King-ho Cheung. 2025. "From Adipose to Action: Reprogramming Stem Cells for Functional Neural Progenitors for Neural Regenerative Therapy" International Journal of Molecular Sciences 26, no. 14: 6599. https://doi.org/10.3390/ijms26146599

APA Style

Peng, J., Zhang, Z., Li, M., Yung, K. K. L., & Cheung, K.-h. (2025). From Adipose to Action: Reprogramming Stem Cells for Functional Neural Progenitors for Neural Regenerative Therapy. International Journal of Molecular Sciences, 26(14), 6599. https://doi.org/10.3390/ijms26146599

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