Skip to main content

Cell-based regenerative and rejuvenation strategies for treating neurodegenerative diseases

Abstract

Neurodegenerative diseases including Alzheimer’s and Parkinson’s disease are age-related disorders which severely impact quality of life and impose significant societal burdens. Cellular senescence is a critical factor in these disorders, contributing to their onset and progression by promoting permanent cell cycle arrest and reducing cellular function, affecting various types of cells in brain. Recent advancements in regenerative medicine have highlighted “R3” strategies—rejuvenation, regeneration, and replacement—as promising therapeutic approaches for neurodegeneration. This review aims to critically analyze the role of cellular senescence in neurodegenerative diseases and organizes therapeutic approaches within the R3 regenerative medicine paradigm. Specifically, we examine stem cell therapy, direct lineage reprogramming, and partial reprogramming in the context of R3, emphasizing how these interventions mitigate cellular senescence and counteracting aging-related neurodegeneration. Ultimately, this review seeks to provide insights into the complex interplay between cellular senescence and neurodegeneration while highlighting the promise of cell-based regenerative strategies to address these debilitating conditions.

Introduction

The natural process of growing older involves a gradual deterioration of bodily functions, which significantly increases an organism’s vulnerability to various age-related illnesses, particularly disorders affecting the nervous system. Neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and Amyotrophic lateral sclerosis (ALS), among others, have a substantial impact on an individual’s quality of life and pose a significant burden on society [1]. These neurodegenerative diseases share common underlying mechanisms, including protein aggregation, oxidative stress, inflammation, apoptosis, and mitochondrial dysfunction, all of which contribute to neuronal loss [2]. Cellular senescence plays a crucial role in neurodegenerative disorders and is considered a potential underlying cause. Senescent cells contribute to the onset and advancement of these diseases through various mechanisms [3]. As a key indicator of aging, cellular senescence is defined by permanent cell cycle arrest and a reduction in cellular functions [4]. This process is not limited to somatic cells, but also encompasses stem cells, including neural stem cells (NSCs). When these cells age, their regenerative capacity -essential for maintaining tissue homeostasis and repair- declines [5].

In regenerative medicine, there are three major therapeutic categories known collectively as the “R3” paradigm [6, 7]: (1) Rejuvenation—restoring the functional capacity of existing cells or reversing cellular aging processes; (2) Regeneration—stimulating repair or regrowth of tissues using stem cells or host repair mechanisms; and (3) Replacement—directly substituting lost or damaged cells with functional ones.

The objective of this review is twofold: (1) to critically analyze the processes of cellular senescence that contribute to neurodegenerative disorders, and (2) to discuss cell-based strategies in the R3 context. In recent years, remarkable advancements have been made in the field of regenerative medicine. By integrating R3 concepts, we distinguish how certain approaches focus on rejuvenation, some on regeneration, and others on outright replacement. These strategies aim to counteract the effects of aging and mitigate neurodegeneration by specifically targeting the underlying mechanisms of aging, such as cellular senescence.

This review comprehensively examines the mechanisms of cellular senescence and explores potential R3 strategies. Specifically, it summarizes the role of cellular senescence in neurodegenerative diseases, highlighting its contributions to disease onset, progression acceleration, and the hindrance of traditional treatment effectiveness. Additionally, various cell-based strategies, such as stem cell therapy, direct lineage reprogramming, and partial reprogramming, are explored. Their potential benefits and challenges in treating neurodegenerative diseases are evaluated, with a focus on how these strategies may target senescent cells to restore functionality (rejuvenation), enhance endogenous repair (regeneration), or replace lost neurons (replacement). By delving into these underlying mechanisms and investigating innovative therapeutic approaches, we aim to pave the way for more effective treatments that can enhance patients’ quality of life and potentially delay or even reverse the aging process.

Cellular senescence and neurodegenerative diseases

The association between cellular senescence and neurodegenerative diseases

In neurodegenerative diseases such as AD, PD, and ALS, various cell types within the brain undergo aging-related changes that play a crucial role in initiating and driving disease progression. Neurons, glial cells (including microglia and astrocytes), and NSCs all exhibit senescence-like phenotypes during both physiological aging and neurodegeneration [8]. These changes contribute to the complex pathogenesis of these disorders (Fig. 1).

Fig. 1
figure 1

Impact of cellular senescence and inflammation on neurodegeneration. This figure depicts the progression of neurodegenerative diseases driven by cellular senescence. Key processes include the activation of senescent markers (p65, p53, p16, p21) in astrocytes, which release senescence-associated secretory phenotype (SASP) factors (TNFα, iNOS, IL-1β, IL-6, IL-12, IL-23) that intensify neuroinflammation. Blood-brain barrier (BBB) breakdown leads to neutrophil and leukocyte infiltration, further contributing to inflammation. Microglia activation and the presence of senescent oligodendrocytes result in additional neuronal damage, characterized by Aβ plaque deposition, tau hyperphosphorylation, and myelin degradation, all of which are hallmarks of neurodegeneration

Neurons

Neurons, which have limited regenerative capacity, are highly susceptible to aging-related damage. Interestingly, although neurons were traditionally regarded as exempt from senescence due to their post-mitotic nature, recent evidence shows that they can in fact enter senescence in response to multiple stressors such as oxidative stress and DNA damage [9, 10]. For instance, the loss of SATB1, a DNA binding protein associated with PD, has been shown to activate a cellular senescence transcriptional program in dopaminergic neurons, suggesting neuronal senescence as a contributing factor to PD pathology [11]. Senescent neuronal cells accumulate with age and exhibit characteristics such as cell cycle arrest, pro-inflammatory secretory phenotype, and altered proteostasis, which can exacerbate neuroinflammation and oxidative stress, leading to cognitive decline and neuronal degeneration [12, 13]. This neuronal senescence phenomenon has led to increased interest in senolytic therapies specifically targeting neurons. Such therapies aim to selectively eliminate senescent neurons or modulate their harmful effects on neuronal function [14, 15]. Preliminary studies have shown promising results, with senolytic treatments blocking disease progression in mouse models of tau - mediated neurodegeneration by acting on affected neurons and improving outcomes in SARS-CoV-2-induced neuropathology involving neuronal damage [14, 15]. Nonetheless, further research is needed to thoroughly investigate the occurrence of neuronal senescence and evaluate the long-term safety and efficacy of senolytic treatments in neurodegenerative diseases [16].

Glia cells

Glia cells, particularly microglia and astrocytes, are crucial components of the central nervous system (CNS) that significantly contribute to the pathogenesis of neurodegenerative diseases. Microglia, the primary immune cells of the CNS, undergo age-related and senescence-driven changes that profoundly impact the onset and progression of these disorders [16]. In PD, for example, reactive microglia have been observed in the substantia nigra and striatum, accompanied by elevated levels of pro-inflammatory cytokines [17]. These changes in microglial function can lead to chronic neuroinflammation, exacerbating neuronal damage and accelerating disease progression.

Astrocytes, once considered merely supportive cells, are now recognized as active participants in various complex CNS functions. Like microglia, astrocytes also experience age-related changes that can influence the development and progression of neurodegenerative diseases [17]. These changes may include alterations in astrocyte morphology, reactivity, and their ability to provide metabolic support to neurons. Astrocytes are also integral to maintaining the integrity and function of the blood-brain barrier (BBB), forming part of the neurovascular unit along with brain microvascular endothelial cells (BMVECs), pericytes, and neurons [18]. Senescent astrocytes significantly contribute to neurodegenerative processes and linked to cognitive decline [19, 20]. The accumulation of senescent astrocytes in the aging brain, characterized by lamin-B1 reduction and nuclear deformations, may lead to astrocyte dysfunctions and subsequent neurodegeneration [21]. Their ability to support neuronal survival, clear neurotransmitters, and remove toxic protein aggregates, which are essential for maintaining a healthy CNS environment, is reduced [19]. Interestingly, senescent astrocytes display a unique transcriptome distinct from reactive astrocytes, with dysregulated pathways and downregulation of brain-expressed genes involved in neuronal development and differentiation [22, 23]. Contradictory to their normal protective role, senescent astrocytes exhibit impaired astrocytic responses to injury and decreased expression of genes involved in antigen processing and presentation [22]. Targeting senescent astrocytes and their associated pathways presents a promising approach for developing therapies to counter age-related neurodegeneration and improve brain health [24, 25].

The accumulation of senescent cells in the brain, including oligodendrocytes, contributes to age-related pathologies and neurodegeneration [9]. Oligodendrocytes, specialized glial cells in the CNS, are crucial for maintaining neuronal health and function, not just for myelination. Recent evidence shows that oligodendrocyte dysfunction, such as senescence, significantly contributes to neurodegenerative processes. Senescent oligodendrocytes can produce SASPs, fostering chronic inflammation and oxidative stress, which lead to progressive CNS demyelination, microglial inflammation, and further neurodegeneration [26]. Importantly, the finding that deletion of the p21CIP1 pathway ameliorated the disease, while blocking microglial inflammation did not prevent neurodegeneration in demyelinating disorders suggests that senescence, particularly of oligodendrocytes, is a key factor in driving neurodegeneration. This contradicts the traditional view that microglial inflammation is the sole primary driver and highlights the significance of directly targeting senescent oligodendrocytes as potential therapeutic targets [26].

Pericytes

Senescent pericytes also play a significant role in neurodegenerative processes, contributing to brain aging and neurological disorders. Pericytes, located within the neurovascular unit, are crucial for maintaining BBB integrity, regulating cerebral blood flow, and supporting overall brain health [27, 28]. As pericytes become senescent, they exhibit characteristic features such as increased β-galactosidase activity, cell cycle arrest, and enhanced expression of SASP factors [29]. These changes can lead to BBB dysfunction, reduced cerebral blood flow, and impaired clearance of toxic cellular by-products [30]. Understanding the molecular mechanisms of pericyte senescence and its impact on the neurovascular unit is essential for developing targeted therapies to combat neurodegenerative disorders and enhance brain health during aging.

Neural stem cells (NSCs)

NSCs are pivotal for brain function and maintenance, yet their senescence is closely linked to neurodegenerative processes. Amyloid-β (Aβ), a pathogenic protein associated with AD, has been shown to accelerate cellular senescence in human NSCs [31]. This process involves enhanced expression of senescence-related genes, increased senescence-associated-β-galactosidase (SA-β-gal) activity, and activation of the DNA damage response [31]. Interestingly, reprogramming of human fibroblasts into induced NSCs (iNSCs) using miR-302a has demonstrated promising results in combating cellular aging and improving cognitive performance in AD models [32]. These miR-302a-hiNSCs showed delayed aging, increased resistance to oxidative stress, and improved cognitive function when implanted into senescence-accelerated mice [32]. This contradicts the notion that all NSCs are susceptible to senescence and suggests that certain reprogramming techniques may offer therapeutic potential.

Endothelial cells

Senescent endothelial cells significantly influence neurodegenerative processes by contributing to age-related vascular and neurological disorders. The accumulation of these cells can lead to endothelial dysfunction, which is associated with conditions such as atherosclerosis, diabetes mellitus, hypertension, and ischemic injury [33]. Senescent endothelial cells display a distinct SASP, characterized by the release of pro-inflammatory cytokines, chemokines, and proteases that disrupt the local microenvironment. Unlike in normal, healthy endothelial cells, the SASP in senescent ones initiates a cascade of events that can lead to tissue dysfunction and further exacerbate the aging process at the local level. This unique secretory behavior of senescent endothelial cells thus becomes a crucial focus when studying the mechanisms underlying age-related pathologies and potential therapeutic interventions [34]. For example, exposure to neurotoxic amyloid β (Aβ1–42) oligomers can induce a senescence phenotype in human brain microvascular endothelial cells (HBMECs) [35]. This finding suggests a direct link between endothelial cell senescence and neurodegenerative diseases such as AD. Furthermore, senescent endothelial cells show alterations in morphological and nanomechanical properties, including increased membrane stiffness and changes in adhesion properties, which may contribute to cerebrovascular dysfunction [35].

The challenges and limitations of traditional treatments for neurodegenerative diseases in the context of cellular senescence

Cellular senescence poses significant challenges to traditional treatments for neurodegenerative diseases. It involves multiple aspects that complicate the treatment process.

Senescent cell accumulation, apoptosis resistance, and SASP

The accumulation of senescent cells, their resistance to apoptosis, and the SASP are major issues. The SASP exacerbates neurodegeneration by creating a pro-inflammatory milieu that affects neighboring healthy cells. This not only leads to chronic inflammation and tissue dysfunction but also interferes with treatment efficacy. Senescent cells may respond differently to medications, impair neurogenesis, disrupt the BBB, and exhibit mitochondrial dysfunction. Moreover, senescence-associated epigenetic changes, cellular heterogeneity in affected brain regions, and reduced regenerative capacity of NSCs further compound the challenges in targeted treatment.

Drug delivery and uptake

In terms of drug delivery and uptake, cellular senescence causes changes in membrane permeability and the function of transport proteins. These cells have reduced membrane permeability, affecting drug uptake and efficacy. Altered endocytic activity in these cells, as seen in the blockage of clathrin-dependent receptor-mediated endocytosis in senescent human diploid fibroblasts, impacts their ability to internalize drugs [36]. However, this reduced permeability can also be exploited for targeted drug delivery. Muñoz-Espín et al. describes an innovative approach that takes advantage of the high lysosomal β-galactosidase activity in senescent cells to design a drug delivery system using galacto-oligosaccharides [37]. This method allows for preferential release of encapsulated drugs within senescent cells, improving efficacy and reducing side effects in non-senescent cells [37]. Additionally, changes in transport proteins can lead to altered drug distribution within the cell, resulting in suboptimal drug concentrations at the target sites [38]. Studies have shown that replicative senescence in renal proximal tubular epithelial cells (RPTECs) leads to changes in the expression of various transporters. Notably, the mRNA level of organic cation transporter 2 decreased rapidly with increasing passage numbers, indicating a significant impact of senescence on transporter expression [39]. Furthermore, the uptake of fluorescent cationic substrates was reduced in SA-β-gal-positive RPTECs compared to SA-β-gal-negative cells, suggesting altered transporter function in senescent cells [39]. Besides, the regulation of drug transporter expression and activity can occur at various levels, including transcription, mRNA stability, translation, and post-translational modification, which may be influenced by the senescence process [40].

Drug-target interaction

Regarding drug-target interaction, the SASP can modify the expression or function of target proteins, interfere with intended drug-target interactions, and contribute to chronic inflammation and tissue dysfunction, potentially reducing the effectiveness of conventional treatments. Inflammatory cytokines and chemokines released as part of the SASP can alter signaling pathways, gene expression programs, and even cause conformational changes in drug targets like receptors or enzymes, making it difficult for drugs to bind effectively (41,42, 43). Traditional treatment approaches for neurodegenerative diseases have limitations. These treatments often focus on alleviating symptoms or targeting specific aspects of the disease mechanism rather than addressing cellular senescence, the root cause. For instance, neurotransmitter-enhancing drugs for AD patients only provide symptomatic relief without tackling neuronal senescence issues [3, 44].

Disease heterogeneity

Disease heterogeneity is another challenge. Neurodegenerative diseases are highly heterogeneous in affected cell types and underlying molecular mechanisms, and cellular senescence exacerbates this. Different cell types in the brain, such as glial cells, endothelial cells, neural stem cells, and neurons, can exhibit various senescence-like phenotypes and respond differently to treatments and interventions [8]. This heterogeneity is further complicated by the fact that senescent cells may respond differently to genetic and pharmacological interventions, known as senolytics or senomorphics [45]. Recent advancements, such as the machine learning program SenCID, have enabled the identification of six major senescence identities (SIDs) with different baselines, stemness, gene functions, and responses to senolytics [46]. This improved understanding of senescent cell heterogeneity may contribute to the effective treatments.

Lack of long-term efficacy

There is a lack of long-term efficacy in traditional treatments. Many traditional therapies, like those for ALS, have limited long-term benefits. For example, Riluzole, approved in the 1990s, remains one of the few FDA-approved treatments for ALS, offering only modest survival benefits. More recently, edaravone has been approved, but the search for effective therapies targeting disease progression is still desperately needed [47]. This lack of long-term efficacy highlights the need for more innovative treatment strategies that can address the underlying causes of cellular senescence and disease progression.

Cell-based R3 strategies for treating neurodegenerative disease

In this section, we discuss how “R3” approaches (Rejuvenation, Regeneration, and Replacement) can be applied to mitigate cellular senescence and aid in treating neurodegenerative disorders [48,49,50]. Specifically, we highlight stem cell therapy and cellular reprogramming (direct lineage reprogramming and partial reprogramming) as representative R3 modalities. These strategies aim to mitigate the effects of cellular senescence by (1) rejuvenating existing cells to restore youthful function, (2) regenerating neural tissues through stimulating or transplanting stem cells, or (3) replacing entirely lost neuronal populations with new, functional cells (Fig. 2).

Fig. 2
figure 2

Overview of cell-based strategies for treating neurodegenerative diseases. This diagram summarizes cell-based strategies to treat neurodegenerative diseases. Stem cells, including pluripotent stem cells (PSCs), mesenchymal stem cells (MSCs), and NSCs, may rejuvenate existing cells and regenerate or replace damaged ones by serving as sources for generating neural cell types like neurons, astrocytes, and oligodendrocytes. Non-neuronal cells can also be directly reprogrammed into neurons, bypassing the stem cell stage. Additionally, aged neural cells undergo transient reprogramming to a stem-like state, promoting rejuvenation and potentially reversing age-related damage. These methods aim to produce functional neural cells that could replace or repair damaged cells in neurodegenerative conditions, helping to restore brain function and slow disease progression

Stem cell therapy

Stem cells are characterized by two key features: self-renewal, which allows them to divide repeatedly; potency, which enables them to differentiate into various specialized cell types 5152, 53. Based on these properties, stem cells are categorized into totipotent, pluripotent, multipotent, or unipotent [51, 54]. Most mature human cells are highly differentiated, unable to divide, and are known as “post-mitotic cells” [55]. When these differentiated post-mitotic cells become senescent or die due to natural aging, injury, or disease, their regeneration depends on the activation of local stem cells, which replenishes the tissue with younger, healthy cells [56]. However, in the aging brain or neurodegenerative conditions, local stem cells often lose their regenerative potential [57]. Consequently, stem cell therapy has emerged as a promising R3 approach for treating neurodegenerative conditions [58,59,60,61], by using stem cells or stem cell-derived neurons/NSCs to repair or replace damaged and senescent neurons, thereby supporting neural function, regeneration, and rejuvenation (Table 1).

Pluripotent stem cells (PSCs)

Cell sources

PSCs are a type of stem cell capable of differentiating into any cell type in the body, making them highly valuable for therapeutic applications. PSCs can be derived from various sources, including embryonic stem cells (ESCs) [62] and induced pluripotent stem cells (iPSCs)63, 64 65, 66. ESCs originate from the inner cell mass of blastocysts but raise ethical concerns due to embryos destruction [62]. iPSCs are generated by reprogramming adult somatic cells (e.g., skin fibroblasts) through transcription factors (OCT4, SOX2, KLF4, and c-MYC) [63, 64] or chemical compounds [65, 66] that reset the cells’ developmental clock, sharing many ESC-like properties while avoiding ethical issues. They allow for patient-specific therapies, as they can be created from the patient’s own cells, reducing the risk of immune rejection.

Neural cell differentiation from PSCs

Efficient differentiation from PSCs into neurons is critical for stem cell replacement therapy against neurodegenerative diseases. In this process, PSCs are induced into neuronal progenitors and further specified into specific neuronal subtypes using factors like brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), and others, tailored to produce dopaminergic (DA) neurons for PD treatment [67,68,69], cholinergic neurons for AD treatment [70, 71], or motor neurons for ALS treatment [72, 73].

PSCs-based therapy

PSC-based therapies can function within the replacement dimension of the R3 paradigm by transplanting PSC-derived specialized neurons that restore lost neural functions and counteract disease progression [74]. In some cases, they may also promote rejuvenation or regeneration via paracrine signaling and local microenvironment modulation.

PD

In 2011 and 2017, pre-clinical studies established proof of concept for dopamine (DA) neurons derived from human ESCs (hESCs) and human induced PSCs (hiPSCs) [75, 76]. Later then, more pre-clinical studies reported that PSC-based therapy can contribute to the treatment of PD [77,78,79,80,81,82,83,84,85]. In 2018, a clinical trial commenced involving the surgical transplantation of allogeneic hiPSC-derived DAergic neuron precursors into the putamen of individuals with PD [86, 87]. In 2020, a study documented the clinical implantation of patient-derived midbrain dopaminergic (mDA) progenitor cells, differentiated in vitro from autologous iPSCs, in a patient with idiopathic PD [88]. Ongoing clinical trials involving PSCs for PD have shown promise in terms of safety and efficacy [84, 85, 89, 90].

AD

Recent animal model studies have shown potential for PSC-based therapies to reduce pathology and enhance cognitive function [91,92,93,94,95]. Transplanting NSCs derived from mouse adult brains or PSCs into AD rodent models led to the generation of cholinergic neurons, increased synaptic strength, and improved memory performance [96, 97]. Additionally, hiPSC-derived NPCs and cholinergic neurons successfully survived and differentiated into cholinergic and GABAergic neurons in the host brain, leading to improved spatial memory [98,99,100].

ALS

Although iPCS is increasingly used in ALS research, the studies published so far have mainly focused on iPSC-based ALS cellular models for disease mechanisms or drug screening [101,102,103]. Some studies utilizing iPSC-derived NSC transplants in ALS mouse models have demonstrated positive therapeutic effects, including extended lifespan, enhanced neuromuscular function, and neuroprotection [104,105,106,107,108].

Despite the promise, PSC-based therapies face hurdles in aging-related contexts. One significant concern is that aberrant DNA methylation patterns and histone modifications, which may be exacerbated by senescent cellular environments, can lead to improper differentiation of PSCs into desired neuronal subtypes or reduce the functionality of the resulting cells [109]. Furthermore, tumorigenicity remains a critical risk, as PSCs can form teratomas if not carefully monitored. Advanced imaging techniques and biomarkers are being developed to detect early signs of teratoma formation, but the risk persists during pre-clinical and clinical trials [110]. Immune rejection is another hurdle, as PSCs may elicit an immune response. Researchers are exploring modifications to the epigenetic and genetic profiles of PSCs during reprogramming to reduce this risk [111, 112]. Moreover, functional integration into existing neural networks is challenging in an aged or degenerating brain. Studies suggest that applying specific electrical fields can enhance synapse formation between transplanted neurons and native cells [113, 114], but achieving consistent and effective integration remains a challenge within the context of neurodegenerative diseases.

Mesenchymal stem cells (MSCs)

Cell sources

MSCs are multipotent stromal cells derived from various tissues, including bone marrow, adipose tissue, and umbilical cord blood [115], and more recently, dental pulp. Bone marrow is the original and most extensively studied source [116], known for its well-characterized MSCs. Adipose tissue provides a more accessible and abundant source [117], with less invasive extraction procedures compared to bone marrow. The umbilical cord is a non-invasive source that yields MSCs with high proliferative capacity and immunomodulatory properties [118], while the placenta and amniotic fluid are rich in MSCs and offer potent regenerative properties in an ethically favorable context [119]. Dental pulp-derived MSCs have shown unique properties and promising results in pre-clinical studies for neurodegenerative diseases [120], demonstrating enhanced neuroprotective and regenerative capabilities compared to MSCs from traditional sources.

MSCs-based therapy

MSCs have the capacity to differentiate into a variety of cell types such as osteoblasts, chondrocytes, adipocytes, and even neurons [121]. They show potential for mitigating senescence and promoting rejuvenation through the secretion of anti-inflammatory and neurotrophic factors, as well as exosomes [122,123,124,125]. Recent studies have underscored MSCs’ potential to enhance cognitive function and neuronal health in age-related neurodegenerative diseases [126, 127].

AD

In AD models, MSCs have demonstrated the ability to reduce amyloid-beta plaques, accompanied by improvements in cognitive functions, as observed in pre-clinical studies [128,129,130,131,132]. A Phase I clinical trial led by Kim et al. involved nine patients with mild-to-moderate AD, resulting in stabilization of cognitive decline and enhancement of cognitive function [129, 133, 134].

PD

In the context of PD, MSCs have been utilized to protect and preserve DA neurons. Pre-clinical studies have demonstrated that MSC transplantation can enhance motor function [135], attributed to their neuroprotective properties and their potential to differentiate into neuron-like cells that can assimilate into existing neural pathways [136]. Moreover, a 2021 Phase I clinical trial studied intravenous infusion of umbilical cord MSCs (UC-MSCs) for PD treatment [137].

ALS and MS

MSC therapy may also help mitigate inflammation and demyelination in ALS and MS [138, 139]. In ALS animal models, MSCs have decreased inflammation and promoted remyelination, improving neurological function [140, 141]. These promising animal study results have catalyzed clinical trials assessing MSC therapy’s effects in ALS patients, with early studies indicating safety and therapeutic benefits, including symptom stabilization and improvements in motor abilities and quality of life [142,143,144,145].

Despite their promise in aging-related neurodegenerative diseases, several challenges remain. In an aging body, ensuring the long-term safety and consistent efficacy of MSC therapies is of utmost importance. For instance, as cells age, there may be an increased risk of abnormal cellular behaviors, and potential risks such as tumor formation associated with MSC therapies must be carefully addressed [146, 147]. Developing standardized protocols for the isolation, expansion, and administration of MSCs is highly necessary in the context of aging. The aging process can affect the characteristics of MSCs, making it essential to have consistent procedures to ensure reproducibility and efficacy across studies [148, 149]. Moreover, extensive clinical trials designed to meet regulatory standards are needed before MSC-based therapies gain wide clinical acceptance, particularly in complex aging condition like neurodegeneration.

Neural stem cells (NSCs)

Cell sources

NSCs has gained significant attention over the past few decades as a potential treatment for neurodegenerative disease [150]. They are multipotent cells capable of differentiating into neurons, astrocytes, and oligodendrocytes. NSCs can be obtained for therapeutic purposes through several methods: isolation from fetal or adult brain tissue where NSCs naturally reside [151], differentiation from PSC [152], and direct reprogramming of somatic cells into NSCs by introducing specific TFs or chemical compounds [153].

NSCs-based therapy

In the R3 paradigm, NSC-based therapy primarily fosters regeneration and replacement by generating new neurons and glial cells, which helps rejuvenate the brain. Specifically, NSCs replace aged or damaged cells, promote a healthy neural environment, and enhance neurogenesis to counteract the decline linked with aging and neurodegeneration [154,155,156,157]. Pre-clinical studies of NSCs-based therapy in animal models have shown promising results, with NSCs promoting neural repair, reducing inflammation, and improving function in conditions like ALS, PD and AD [150].

ALS&MS

In mouse models of ALS, implanted human NSCs have been effective in delaying disease onset and progression and increasing survival by producing neurotrophic factors and reducing neural inflammation [158,159,160]. The FDA approved the first Phase I clinical trial in 2009 for transplanting human spinal cord-derived NSCs into ALS patients, followed by a Phase II trial [161,162,163]. These trials, which involved transplanting NSCs into the spinal cords of 6 and 18 ALS patients, respectively, did not show serious adverse reactions or accelerated disease progression [164, 165]. Clinical trials have also explored allogeneic NSC transplantation in patients with progressive MS, demonstrating that the procedure is well-tolerated, feasible, and safe, and it helped slow disease progression [162, 166, 167]. However, other trials have noted potential side effects, such as acute neurological deterioration and central pain syndrome [168].

PD

Recent studies reveal NSC secretions can protect PD neurons, improving both motor and non-motor functions and preserved dopaminergic neurons in PD rats [169,170,171]. In PD animal models, NSC or neural progenitor cell transplants have consistently shown positive therapeutic outcomes [172,173,174]. In PD mice, NSCs that express dopamine-related markers increased astrocyte and microglia presence and enhanced non-phosphorylated neurofilaments in the motor cortex, improving hyperactivity and gait [175]. In clinical trials, human neural progenitor cells/NSCs were transplanted into the dorsal putamen of eight patients with moderate PD, proving safe without immune or adverse reactions, and most patients showed motor improvements [176, 177]. Another trial involving NSC transplants into the striatum of 21 PD patients showed significant symptom improvement with no side effects, such as tumor formation or immune rejection [178].

AD

NSC transplantation is a promising therapeutic approach for AD, as NSCs can differentiate into cholinergic neurons, thus replacing and improving lost neurons [179]. They also enhance neuronal connectivity and metabolic activity in AD through compensatory mechanisms, preserving cognitive function [180, 181]. NSCs have also been shown to reduce amyloid-beta plaques, either through direct interaction or by modulating the immune response [179].

Despite these promising aspects, several challenges remain. Key issues include ensuring the long-term survival and integration of transplanted cells, preventing tumorigenesis, and understanding the complex interactions between NSCs and host brain tissue [150]. Ongoing advancements in NSC differentiation protocols, delivery methods, and safety measures aim to address these challenges. Overall, NSC therapy provides a comprehensive approach to treating neurodegenerative diseases by supporting rejuvenation and combating senescence.

Table 1 Summary of stem cell therapy for treating neurodegenerative disease

Potential mechanisms of stem cell therapies

The underlying mechanisms of stem cell therapy are complex and multifaceted, encompassing neuronal replacement, paracrine mechanisms, immune modulation, neurotrophic support, mitochondrial transfer [183, 184] (Fig. 3).

Fig. 3
figure 3

Potential mechanisms underlying the stem cell therapies. This schematic illustrates how stem cell–based therapies combat neurodegeneration through multiple mechanisms in one integrated approach. Newly grafted or converted neural cells (center) replace or supplement degenerating neurons (upper left), while transplanted stem cells or induced cells also bolster neurogenesis (upper right) by differentiating into neurons, astrocytes, oligodendrocytes, or interneurons. Moreover, these cells secrete neurotrophic factors (left) that enhance cell survival and plasticity, and release anti-inflammatory mediators (lower left) to modulate immune responses involving peripheral macrophages, microglia, and astrocytes. Finally, healthy mitochondria can be transferred to damaged neurons (lower right), improving energy production and reducing oxidative stress

Neuronal replacement

One of the primary mechanisms by which stem cell therapy operates is through neuronal replacement (the “replacement” aim of the R3 paradigm) [185,186,187], thereby addressing the fundamental problem of neuronal loss and aging. In PD, for instance, stem cells can become dopaminergic neurons, integrating into existing neural circuits to restore functions [75, 188,189,190,191]. Similarly, hESC-derived neurons can improve motor function in Huntington’s disease models [192].

Neurogenesis

Stem cell therapies offer regeneration potential by promoting neurogenesis, which is the generation of new neurons from neural stem cells. During neurogenesis, transplanted stem cells migrate to areas like the hippocampus, vital for memory and learning, where they replenish the neuronal population by generating new neurons [193]. Interestingly, stem cell therapy can support the activity of endogenous NSCs, enhancing neurogenesis by creating a favorable environment and secreting factors that promote neural differentiation and growth [132, 193].

Neurotrophic support

Stem cells contribute significantly to neurotrophic support by producing and releasing factors like BDNF and GDNF [108, 182, 194, 195] and other neurotrophic factors [196, 197]. Administration of MSC-overexpressing neurotrophic factors such as GDNF, vascular endothelial growth factor (VEGF), BDNF into mouse model of PD [198], AD [199, 200] and ALS [201] resulted in promising in vivo outcomes. This neurotrophic support aligns with the rejuvenation aspect of R3, preserving neurons and delay senescence of both neurons and glial cells, thereby enhancing overall brain health and longevity [202, 203].

Anti-neuroinflammation

Neuroinflammation involves various chronic, pro-inflammatory, immune system-mediated processes, mostly allied with neurodegeneration [204,205,206]. Stem cells modulate immune responses by dampening neuroinflammation [207]. In a transgenic mouse model of ALS, administration of MSCs could hinder disease progress by downregulating inflammatory inducible nitric oxide synthase (iNOS) activation [208] and suppressing expression of pro-inflammatory cytokines in vivo [208, 209]. This immunomodulatory effect is critical for rejuvenation, as it reduces the burden of the SASP, which is associated with chronic inflammation and contributes to the perpetuation of neurodegenerative processes [210].

Mitochondrial transfer

Mitochondrial dysfunction is a key factor in the progression of many neurodegenerative diseases, and the ability of stem cells to enhance mitochondrial function in damaged neurons represents a significant therapeutic advantage [211,212,213]. For instance, restoring mitochondrial function in aged NSCs with Piracetam can improve hippocampal neurogenesis in vivo in old mice [214]. In some studies, MSCs have been shown to transfer healthy mitochondria to damaged neurons, which is crucial for restoring cellular energy production and reducing oxidative stress [215, 216]. By improving mitochondrial function, stem cells support the rejuvenation dimension of the R3 paradigm, helping delay cellular senescence and maintain the functional integrity of neurons, thus supporting the overall goal of brain rejuvenation [217, 218].

Cellular reprogramming

Direct lineage reprogramming

Direct lineage reprogramming, also known as trans-differentiation, is a transformative process that converts one differentiated cell type directly into another without passing through a pluripotent stem cell stage [219, 220]. In the context of neuroscience, this method involves converting non-neuronal cells directly into neurons [221], offering promising research and therapeutic applications, particularly as regenerative strategies for neurodegenerative diseases [222].

Genetic and chemical reprogramming

Genetic methods typically involve overexpressing specific TFs or microRNAs to induce neuronal identity. For example, a combination of Ascl1, Brn2, and Myt1l can convert fibroblasts or hepatocytes into functional neurons, known as induced neurons (iNs) [223, 224]. Similarly, Other TF combinations can reprogram fibroblasts and astrocytes into neurons or neural stem cells (NSCs) [225,226,227,228]. As for chemical methods, chemical cocktails consisting of Forskolin, CHIR99021, I-BET151, and Wnt/β-catenin agonists efficiently convert mouse fibroblasts to neurons [229]. A cocktail of CHIR99021 and other molecules has been demonstrated to reprogram fibroblasts into NSCs [230, 231]. Human fibroblasts were converted to vGLUT1-positive glutamatergic neurons using a combination of specific chemical compounds [232]. Astrocytes could also be converted to neurons through chemical reprogramming [233,234,235].

In vitro and in vivo reprogramming

In vitro reprogramming involves converting non-neuronal cells, such as fibroblasts or glial cells, into neurons within a controlled laboratory environment. Different neuronal subtypes, including dopaminergic neurons [226, 236, 237], glutamatergic neurons [238,239,240] and motor neurons [241], have been generated in vitro. This capability is useful for creating patient-specific neurons for cell replacement therapies. In vivo reprogramming bypass the need for cell delivery by converting local glial cells to neurons within the brain [242, 243]. This process employs TFs or chemical compounds to reprogram cells at the target site. Commonly used TFs for neural reprogramming include NeuroD1, Ascl1, Brn2, and Myt1l, which together convert glial cells into functional neurons [244, 245]. Chemical reprogramming in vivo leverages small molecules that can modulate signaling pathways, epigenetic states, and transcriptional networks to induce cellular reprogramming, especially astrocytes to neurons [246]. A cocktail named FICBY has demonstrated success in transforming astrocytes into neurons in vivo [235].

Mechanism of action

n line with the R3 paradigm, direct lineage reprogramming supports “Regeneration” or “Replacement” by converting non-neural cells (e.g., glial cells or fibroblasts) into functional neurons. This process allows for neuron regeneration directly at injury sites, promoting repair and restoring function in neurodegenerative diseases while avoiding challenges associated with stem cell transplantation, like cell delivery and integration.

Advantages

Direct lineage reprogramming avoids the risks of tumorigenesis associated with pluripotency in iPSCs or ESCs [219, 220]. In vivo reprogramming allows conversion within the brain’s intrinsic environment, supporting better integration and functionality [247]. It also reduces the need for cell transplantation, lowering the risk of immune rejection. Additionally, since patient-specific cells can be directly reprogrammed into neurons, this technique holds great potential for creating personalized models of neurological diseases.

Challenges

Reprogramming can result in incomplete transformation and integration into neural networks, with potential off-target effects [248]. Ensuring the functionality and maturity of reprogrammed neurons in vitro, is challenging, as is their survival post-transplantation. In vivo methods require precise control to avoid aberrant reprogramming, which could lead to tumorigenesis or other adverse effects [222]. Additionally, the efficiency of reprogramming and long-term survival of newly generated neurons are critical issues that need addressing.

Partial reprogramming

Partial reprogramming, which aligns with the rejuvenation aspect of the R3 paradigm, involves the temporary activation of OSKM factors (OCT4, SOX2, KLF4, and c-MYC) to reverse cellular aging effects, such as telomere shortening, mitochondrial dysfunction, and altered gene expression [207, 249, 250]. This rejuvenation effect is achieved without pushing the cells into a fully pluripotent state, which would erase their identity and lead to potential risks such as tumorigenesis.

Therapeutic potential

Several studies have demonstrated the potential of partial reprogramming to induce rejuvenation across a variety of tissues in mice including pancreas and muscle [251, 252], to different extents depending on the cell type [253,254,255]. In a study, mice subjected to periodic activation of OSKM factors exhibited reduced signs of aging, improved tissue regeneration, and better overall health, suggesting that partial reprogramming can be an effective strategy for promoting rejuvenation [256]. Another study demonstrated that partial reprogramming increased the lifespan of older mice by 109% compared to controls [257].

Partial reprogramming has also emerged as a promising approach for treating neurodegenerative diseases [258, 259], with effects observed in the spinal cord [260] and brain regions such as the optic nerve, hippocampus, and striatum [261,262,263,264]. Improvements have been noted in neuroblast populations in aged neurogenic niches with whole-body partial reprogramming [265]. Another promising application of partial cell reprogramming in brain is restoration of visual function [262]. When inducible OSK-containing AAV9 was delivered to the retinal ganglion cells of old mice via intravenous delivery, continuous expression of OSK factors led to a partially restored vision [262].

Mechanism of action

Partially reprogrammed mice apparently exhibited rejuvenation of certain cellular phenotypes, including the reduction of mitochondrial ROS and restoration of H3K9me levels [249]. Cyclic cell partial reprogramming was shown to return the transcriptome, lipidome, and metabolome of multiple tissues to a younger state [253]. Interestingly, targeting partial reprogramming specifically to the neurogenic niche also boosts the proportion of neuroblasts and their precursors in old mice and improves several molecular signatures of aging, suggesting that the beneficial effects of reprogramming are niche intrinsic [265]. Additionally, in cellular models, partial reprogramming has been shown to rejuvenate aged human cells, making them function more like their younger counterparts [252]. This includes improvements in cellular metabolism, reduced oxidative stress, and enhanced DNA repair mechanisms, all of which contribute to a more youthful cellular phenotype [252].

Benefits and risks

The benefits of partial reprogramming using OSKM factors include potential neuronal regeneration and neuroprotection from degenerative diseases like AD and PD, along with reduced secondary degeneration from neuroinflammation and oxidative stress [252]. However, risks include potential tumorigenesis from improper regulation of pluripotency activation that increase proliferation and suppress somatic cell identity [252, 266, 267], leading to unwanted cell proliferation [268, 269]. Issues such as incomplete reprogramming, coding mutations, and cell heterogeneity pose complications for therapeutic applications [270, 271]. Additionally, unintended differentiation pathways could form non-functional or maladaptive cells, with examples of liver and intestinal failure noted from continuous Yamanaka factor expression in mice [272, 273]. Therefore, robust pre-clinical and clinical evaluations are critical to determine the safety and efficacy of partial reprogramming before its clinical adoption.

Challenges and future directions in treating neurodegenerative diseases

Cell-based strategies rooted in the R3 paradigm (rejuvenation, regeneration, and replacement) hold great promise in regenerative medicine for neurodegenerative diseases. However, several challenges specific to this context need to be overcome.

Limited survival and engraftment in aging context

In the aging population, mechanical stress during implantation, loss of extracellular matrix, and nutrient and oxygen deprivation are exacerbated, along with host inflammatory responses, often leading to significant cell death post-transplantation [274]. This issue is compounded by the age-related decline in adult stem cell functionality, reducing the efficacy of regenerative therapies [275]. The disparity between results in animal models and aged human patients emphasizes the need for more relevant pre-clinical models and a better understanding of age-related stem cell changes [275]. Furthermore, the translation of cellular therapies from “bench to bedside” remains challenging due to various intrinsic and extrinsic barriers, including manufacturing, regulatory, reimbursement, and clinical adoption issues [276].

Safety and efficacy evaluation in neurodegenerative diseases

Comprehensive assessment of safety and efficacy is crucial. While pre-clinical data are promising, clinical trials must determine the true safety and effectiveness in humans with neurodegenerative diseases. Rigorous trial protocols and appropriate outcome measures are required. Long-term follow-up is essential to detect potential late-emerging adverse effects, such as in partial reprogramming, where long-term consequences like tumorigenesis or epigenetic instability need to be ruled out.

Personalized medicine for heterogeneous neurodegenerative diseases

Given the heterogeneity of neurodegenerative diseases, personalized medicine is vital. Biomarkers are needed to predict patient responses. For stem cell therapy, these could be based on genetic profiles, disease stages, and cellular damage levels to select suitable candidates [277]. Similar considerations apply to direct lineage and partial reprogramming to identify patients likely to benefit and those at risk [278,279,280,281].

Regulatory and ethical challenges in aging-related therapies

The development and application of these R3-based strategies raise regulatory and ethical questions [282]. Regulatory requirements for different applications, especially involving stem cell-derived tissue constructs in neurodegenerative disease treatment, are complex and lack clear guidance [283]. Ethical issues include implications for donors, risks related to iPSCs like unwanted differentiation or malignancy, and concerns about MSCs promoting tumor growth, especially in the context of an aging population seeking these therapies [284].

Conclusion

Remarkable progress has been made in R3-focused cell-based strategies for treating neurodegenerative diseases. Advances in stem cell therapy, direct lineage reprogramming, and partial reprogramming offer promising solutions for addressing cellular deficits. Emerging technologies like gene editing and combinatorial therapies provide additional opportunities. However, challenges such as cell and factor delivery and integration, safety and efficacy evaluation, personalized medicine, and regulatory and ethical considerations still exist. Overcoming these challenges requires continued research, collaboration, and innovation. With further progress, R3-based therapies have the potential to transform the treatment of neurodegenerative diseases and bring new hope to patients.

Data availability

All additional files are included in the manuscript.

Abbreviations

AD:

Alzheimer’s Disease

PD:

Parkinson’s disease

ALS:

Amyotrophic lateral sclerosis

RPE:

Retinal pigment epithelium

ASO:

Antisense oligonucleotide

CNS:

Central nervous system

BBB:

Blood brain barrier

BMVECs:

Brain microvascular endothelial cells

SASP:

Senescence-associated secretory phenotype

RPTECs:

Renal proximal tubular epithelial cells

SA-β-gal:

Senescence-associated-β-galactosidase

NSCs:

Neural stem cells

iNSCs:

Induced neural stem cells

iPSCs:

Induced pluripotent stem cells

ESCs:

Embryonic stem cells

PSCs:

Pluripotent stem cells

DA:

Dopaminergic

mDA:

Midbrain dopaminergic

NPCs:

Neural progenitor cells

MSCs:

Mesenchymal stem cells

UC-MSCs:

Umbilical cord mesenchymal stem cells

BM-MSCs:

Bone marrow mesenchymal stem cells

hESCs:

Human embryonic stem cells

hiPSCs:

Human induced pluripotent stem cells

BDNF:

Brain-derived neurotrophic factor

GDNF:

Glial cell line-derived neurotrophic factor

iNs:

Induced neurons

OSKM:

OCT4, SOX2, KLF4, and cMYC

TFs:

Transcription factors

FGF:

Fibroblast growth factor

SHH:

Sonic hedgehog

VEGF:

Vascular endothelial growth factor

iNOS:

Inducible nitric oxide synthase

HBMECs:

Human brain microvascular endothelial cells

Aβ:

Amyloid-β

References

  1. Agnello L, Ciaccio M. Neurodegenerative diseases: from molecular basis to therapy. Int J Mol Sci. 2022;23.

  2. Tarawneh R, Galvin JE. Potential future neuroprotective therapies for neurodegenerative disorders and stroke. Clin Geriatr Med. 2010;26:125–47.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Thapa R, Ahmad Bhat A, Shahwan M, Ali H, PadmaPriya G, Bansal P, Rajotiya S, Barwal A, Siva Prasad GV, Pramanik A, Khan A, Hing Goh B, Dureja H, Kumar Singh S, Dua K, Gupta G. Proteostasis disruption and senescence in Alzheimer’s disease pathways to neurodegeneration. Brain Res. 2024;1845:149202.

    Article  CAS  PubMed  Google Scholar 

  4. de Magalhães JP. Cellular senescence in normal physiology. Science. 2024;384:1300–1.

    Article  PubMed  Google Scholar 

  5. Farahzadi R, Valipour B, Montazersaheb S, Fathi E. Targeting the stem cell niche micro-environment as therapeutic strategies in aging. Front Cell Dev Biol. 2023;11:1162136.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Nelson TJ, Behfar A, Terzic A. Strategies for therapeutic repair: the R(3) regenerative medicine paradigm. Clin Transl Sci. 2008;1:168–71.

    Article  PubMed  PubMed Central  Google Scholar 

  7. McKinley KL, Longaker MT, Naik S. Emerging frontiers in regenerative medicine. Science. 2023;380:796–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhang P, Sung M-H. Chapter 14 - Cellular senescence in neurodegenerative diseases. In: Serrano M, Muñoz-Espín D, editors. Cellular senescence in disease. Academic; 2022. pp. 363–81.

  9. de Luzy IR, Lee MK, Mobley WC, Studer L. Lessons from inducible pluripotent stem cell models on neuronal senescence in aging and neurodegeneration. Nat Aging. 2024;4:309–18.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Walton CC, Andersen JK. Unknown fates of (brain) oxidation or UFO: close encounters with neuronal senescence. Free Radic Biol Med. 2019;134:695–701.

    Article  CAS  PubMed  Google Scholar 

  11. Riessland M, Kolisnyk B, Kim TW, Cheng J, Ni J, Pearson JA, Park EJ, Dam K, Acehan D, Ramos-Espiritu LS, Wang W, Zhang J, Shim JW, Ciceri G, Brichta L, Studer L, Greengard P. Loss of SATB1 induces p21-Dependent cellular senescence in Post-mitotic dopaminergic neurons. Cell Stem Cell. 2019;25:514–e5308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Behfar Q, Ramirez Zuniga A, Martino-Adami PV. Aging, senescence, and dementia. J Prev Alzheimers Dis. 2022;9:523–31.

    Article  CAS  PubMed  Google Scholar 

  13. Martínez-Cué C, Rueda N. Cellular senescence in neurodegenerative diseases. Front Cell Neurosci. 2020;14:16.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Aguado J, Amarilla AA, Taherian Fard A, Albornoz EA, Tyshkovskiy A, Schwabenland M, Chaggar HK, Modhiran N, Gómez-Inclán C, Javed I, Baradar AA, Liang B, Peng L, Dharmaratne M, Pietrogrande G, Padmanabhan P, Freney ME, Parry R, Sng JDJ, Isaacs A, Khromykh AA, Valenzuela Nieto G, Rojas-Fernandez A, Davis TP, Prinz M, Bengsch B, Gladyshev VN, Woodruff TM, Mar JC, Watterson D, Wolvetang EJ. Senolytic therapy alleviates physiological human brain aging and COVID-19 neuropathology. Nat Aging. 2023;3:1561–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mendelsohn AR, Larrick JW. Cellular senescence as the key intermediate in Tau-Mediated neurodegeneration. Rejuvenation Res. 2018;21:572–9.

    Article  CAS  PubMed  Google Scholar 

  16. Spittau B. Aging Microglia-Phenotypes, functions and implications for Age-Related neurodegenerative diseases. Front Aging Neurosci. 2017;9:194.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Kattimani YM, Waisman A. Microglia’s heroic battle against neurodegeneration. Neuron. 2024;112:2661–3.

    Article  CAS  PubMed  Google Scholar 

  18. Gastfriend BD, Palecek SP, Shusta EV. Modeling the blood-brain barrier: beyond the endothelial cells. Curr Opin Biomed Eng. 2018;5:6–12.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Kim D, Yoo SH, Yeon GB, Oh SS, Shin WH, Kang HC, Lee CK, Kim HW, Kim DS. Senescent astrocytes derived from human pluripotent stem cells reveal Age-Related changes and implications for neurodegeneration. Aging Dis. 2024. Epub ahead of print. https://doiorg.publicaciones.saludcastillayleon.es/10.14336/AD.2024.0089

  20. Han X, Zhang T, Liu H, Mi Y, Gou X. Astrocyte senescence and Alzheimer’s disease: A review. Front Aging Neurosci. 2020;12:148.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Matias I, Diniz LP, Damico IV, Araujo APB, Neves LDS, Vargas G, Leite REP, Suemoto CK, Nitrini R, Jacob-Filho W, Grinberg LT, Hol EM, Middeldorp J, Gomes FCA. Loss of lamin-B1 and defective nuclear morphology are hallmarks of astrocyte senescence in vitro and in the aging human hippocampus. Aging Cell. 2022;21:e13521.

    Article  CAS  PubMed  Google Scholar 

  22. Crowe EP, Tuzer F, Gregory BD, Donahue G, Gosai SJ, Cohen J, Leung YY, Yetkin E, Nativio R, Wang LS, Sell C, Bonini NM, Berger SL. Johnson FB and Torres C. Changes in the transcriptome of human astrocytes accompanying oxidative Stress-Induced senescence. Front Aging Neurosci. 2016;8:208.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Simmnacher K, Krach F, Schneider Y, Alecu JE, Mautner L, Klein P, Roybon L, Prots I, Xiang W, Winner B. Unique signatures of stress-induced senescent human astrocytes. Exp Neurol. 2020;334:113466.

    Article  CAS  PubMed  Google Scholar 

  24. Cohen J, Torres C. Astrocyte senescence: evidence and significance. Aging Cell. 2019;18:e12937.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Wang X, Deng Y, Xiao Y, Wang F, Tang Z, Qi X. A double inducible cell ablation system for eliminating senescent astrocytes via apoptosis. Mol Biol Rep. 2024;51:363.

    Article  CAS  PubMed  Google Scholar 

  26. Rivellini C, Porrello E, Dina G, Mrakic-Sposta S, Vezzoli A, Bacigaluppi M, Gullotta GS, Chaabane L, Leocani L, Marenna S, Colombo E, Farina C, Newcombe J, Nave KA, Pardi R. Quattrini A and previtali SC. JAB1 deletion in oligodendrocytes causes senescence-induced inflammation and neurodegeneration in mice. J Clin Invest. 2022;132.

  27. Bell RD, Winkler EA, Sagare AP, Singh I, LaRue B, Deane R, Zlokovic BV. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron. 2010;68:409–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Shimizu F, Kanda T. [Pericytes of the nervous system: physiological and pathological role]. Brain Nerve. 2020;72:151–8.

    CAS  PubMed  Google Scholar 

  29. Iwao T, Takata F, Matsumoto J, Goto Y, Aridome H, Yasunaga M, Yokoya M, Kataoka Y, Dohgu S. Senescence in brain pericytes attenuates blood-brain barrier function in vitro: A comparison of serially passaged and isolated pericytes from aged rat brains. Biochem Biophys Res Commun. 2023;645:154–63.

    Article  CAS  PubMed  Google Scholar 

  30. Winkler EA, Sagare AP, Zlokovic BV. The pericyte: a forgotten cell type with important implications for Alzheimer’s disease? Brain Pathol. 2014;24:371–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Li R, Li Y, Zuo H, Pei G, Huang S, Hou Y. Alzheimer’s Amyloid-β Accelerates Cell Senescence and Suppresses SIRT1 in Human Neural Stem Cells. Biomolecules. 2024;14(2):189. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biom14020189

  32. Li Y, Sun J, Zheng Y, Xu T, Zhang Y, Wang Y. MiR-302-Induced anti-aging neural stem cells enhance cognitive function and extend lifespan. BioRxiv. 2023:2023.02.13.528232.

  33. Yamakuchi M, Hashiguchi T. Endothelial cell aging: how MiRNAs contribute?? J Clin Med. 2018;7.

  34. Wong PF, Tong KL, Jamal J, Khor ES, Lai SL, Mustafa MR. Senescent HUVECs-secreted exosomes trigger endothelial barrier dysfunction in young endothelial cells. Excli J. 2019;18:764–76.

    PubMed  PubMed Central  Google Scholar 

  35. Kulkarni T, Angom RS, Das P, Bhattacharya S, Mukhopadhyay D. Nanomechanical insights: amyloid beta oligomer-induced senescent brain endothelial cells. Biochim Biophys Acta Biomembr. 2019;1861:183061.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Park JS, Park WY, Cho KA, Kim DI, Jhun BH, Kim SR, Park SC. Down-regulation of amphiphysin-1 is responsible for reduced receptor-mediated endocytosis in the senescent cells. Faseb J. 2001;15:1625–7.

    Article  CAS  PubMed  Google Scholar 

  37. Muñoz-Espín D, Rovira M, Galiana I, Giménez C, Lozano-Torres B, Paez-Ribes M, Llanos S, Chaib S, Muñoz-Martín M, Ucero AC, Garaulet G, Mulero F, Dann SG, VanArsdale T, Shields DJ, Bernardos A, Murguía JR. Martínez-Máñez R and Serrano M. A versatile drug delivery system targeting senescent cells. EMBO Mol Med. 2018;10.

  38. Wagner DJ, Hu T, Wang J. Polyspecific organic cation transporters and their impact on drug intracellular levels and pharmacodynamics. Pharmacol Res. 2016;111:237–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Sanagawa A, Hotta Y, Sezaki R, Tomita N, Kataoka T, Furukawa-Hibi Y, Kimura K. Effect of replicative senescence on the expression and function of transporters in human proximal renal tubular epithelial cells. Biol Pharm Bull. 2022;45:1636–43.

    Article  CAS  PubMed  Google Scholar 

  40. Terada T, Inui K. Gene expression and regulation of drug transporters in the intestine and kidney. Biochem Pharmacol. 2007;73:440–9.

    Article  CAS  PubMed  Google Scholar 

  41. Kang C. Senolytics and senostatics: A Two-Pronged approach to target cellular senescence for delaying aging and Age-Related diseases. Mol Cells. 2019;42:821–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Ruscetti M. Abstract SoA2-01: cellular senescence: the senescence-associated secretory phenotype (SASP) and immunity. Cancer Res. 2024;84:SoA2-01-SoA2-01.

  43. Benveniste EN, Cytokines. Encyclopedia of the Neurological Sciences. 2014:921–925.

  44. Tayeb HO, Yang HD, Price BH, Tarazi FI. Pharmacotherapies for Alzheimer’s disease: beyond cholinesterase inhibitors. Pharmacol Ther. 2012;134:8–25.

    Article  CAS  PubMed  Google Scholar 

  45. Lushchak O, Schosserer M, Grillari J. Senopathies-Diseases associated with cellular senescence. Biomolecules. 2023;13.

  46. Tao W, Yu Z, Han JJ. Single-cell senescence identification reveals senescence heterogeneity, trajectory, and modulators. Cell Metab. 2024;36:1126–e11435.

    Article  CAS  PubMed  Google Scholar 

  47. Silani V. Therapy in amyotrophic lateral sclerosis (ALS): an unexpected evolving scenario. Arch Ital Biol. 2017;155:118–30.

    PubMed  Google Scholar 

  48. Wyss-Coray T. Ageing, neurodegeneration and brain rejuvenation. Nature. 2016;539:180–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zocher S. Targeting neuronal epigenomes for brain rejuvenation. Embo J. 2024;43:3312–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ji S, Xiong M, Chen H, Liu Y, Zhou L, Hong Y, Wang M, Wang C, Fu X, Sun X. Cellular rejuvenation: molecular mechanisms and potential therapeutic interventions for diseases. Signal Transduct Target Ther. 2023;8:116.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Res Ther. 2019;10:68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Becker AJ, Mc CE, Till JE. Cytological demonstration of the clonal nature of spleen colonies derived from transplanted mouse marrow cells. Nature. 1963;197:452–4.

    Article  CAS  PubMed  Google Scholar 

  53. Wang X. Stem cells in tissues, organoids, and cancers. Cell Mol Life Sci. 2019;76:4043–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kolios G, Moodley Y. Introduction to stem cells and regenerative medicine. Respiration. 2013;85:3–10.

    Article  PubMed  Google Scholar 

  55. Ruijtenberg S, van den Heuvel S. Coordinating cell proliferation and differentiation: antagonism between cell cycle regulators and cell type-specific gene expression. Cell Cycle. 2016;15:196–212.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Post Y, Clevers H. Defining adult stem cell function at its simplest: the ability to replace lost cells through mitosis. Cell Stem Cell. 2019;25:174–83.

    Article  CAS  PubMed  Google Scholar 

  57. Picerno A, Stasi A, Franzin R, Curci C, di Bari I, Gesualdo L, Sallustio F. Why stem/progenitor cells lose their regenerative potential. World J Stem Cells. 2021;13:1714–32.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Hoang DM, Pham PT, Bach TQ, Ngo ATL, Nguyen QT, Phan TTK, Nguyen GH, Le PTT, Hoang VT, Forsyth NR, Heke M, Nguyen LT. Stem cell-based therapy for human diseases. Signal Transduct Target Ther. 2022;7:272.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Bonaventura G, Munafò A, Bellanca CM, La Cognata V, Iemmolo R, Attaguile GA, Di Mauro R, Di Benedetto G, Cantarella G, Barcellona ML, Cavallaro S, Bernardini R. Stem Cells: Innovative Therapeutic Options for Neurodegenerative Diseases? Cells. 2021;10.

  60. Sivandzade F, Cucullo L. Regenerative stem cell therapy for neurodegenerative diseases: an overview. Int J Mol Sci. 2021;22.

  61. Ford E, Pearlman J, Ruan T, Manion J, Waller M, Neely GG, Caron L. Human pluripotent stem Cells-Based therapies for neurodegenerative diseases: current status and challenges. Cells. 2020;9(11):2517. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells9112517

  62. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145–7.

    Article  CAS  PubMed  Google Scholar 

  63. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76.

    Article  CAS  PubMed  Google Scholar 

  64. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72.

    Article  CAS  PubMed  Google Scholar 

  65. Guan J, Wang G, Wang J, Zhang Z, Fu Y, Cheng L, Meng G, Lyu Y, Zhu J, Li Y, Wang Y, Liuyang S, Liu B, Yang Z, He H, Zhong X, Chen Q, Zhang X, Sun S, Lai W, Shi Y, Liu L, Wang L, Li C, Lu S, Deng H. Chemical reprogramming of human somatic cells to pluripotent stem cells. Nature. 2022;605:325–31.

    Article  CAS  PubMed  Google Scholar 

  66. Hou P, Li Y, Zhang X, Liu C, Guan J, Li H, Zhao T, Ye J, Yang W, Liu K, Ge J, Xu J, Zhang Q, Zhao Y, Deng H. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science. 2013;341:651–4.

    Article  CAS  PubMed  Google Scholar 

  67. Hiller BM, Marmion DJ, Thompson CA, Elliott NA, Federoff H, Brundin P, Mattis VB, McMahon CW, Kordower JH. Optimizing maturity and dose of iPSC-derived dopamine progenitor cell therapy for Parkinson’s disease. NPJ Regen Med. 2022;7:24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Alekseenko Z, Dias JM, Adler AF, Kozhevnikova M, van Lunteren JA, Nolbrant S, Jeggari A, Vasylovska S, Yoshitake T, Kehr J, Carlén M, Alexeyenko A, Parmar M, Ericson J. Robust derivation of transplantable dopamine neurons from human pluripotent stem cells by timed retinoic acid delivery. Nat Commun. 2022;13:3046.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Nakamura R, Nonaka R, Oyama G, Jo T, Kamo H, Nuermaimaiti M, Akamatsu W, Ishikawa KI, Hattori N. A defined method for differentiating human iPSCs into midbrain dopaminergic progenitors that safely restore motor deficits in Parkinson’s disease. Front Neurosci. 2023;17:1202027.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Hong YJ, Do JT. Neural lineage differentiation from pluripotent stem cells to mimic human brain tissues. Front Bioeng Biotechnol. 2019;7:400.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Bissonnette CJ, Lyass L, Bhattacharyya BJ, Belmadani A, Miller RJ, Kessler JA. The controlled generation of functional basal forebrain cholinergic neurons from human embryonic stem cells. Stem Cells. 2011;29:802–11.

    Article  PubMed  Google Scholar 

  72. Sances S, Bruijn LI, Chandran S, Eggan K, Ho R, Klim JR, Livesey MR, Lowry E, Macklis JD, Rushton D, Sadegh C, Sareen D, Wichterle H, Zhang SC, Svendsen CN. Modeling ALS with motor neurons derived from human induced pluripotent stem cells. Nat Neurosci. 2016;19:542–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Workman MJ, Lim RG, Wu J, Frank A, Ornelas L, Panther L, Galvez E, Perez D, Meepe I, Lei S, Valencia V, Gomez E, Liu C, Moran R, Pinedo L, Tsitkov S, Ho R, Kaye JA, Thompson T, Rothstein JD, Finkbeiner S, Fraenkel E, Sareen D, Thompson LM, Svendsen CN. Large-scale differentiation of iPSC-derived motor neurons from ALS and control subjects. Neuron. 2023;111:1191–e12045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Yamanaka S. Pluripotent stem cell-Based cell Therapy-Promise and challenges. Cell Stem Cell. 2020;27:523–31.

    Article  CAS  PubMed  Google Scholar 

  75. Kriks S, Shim JW, Piao J, Ganat YM, Wakeman DR, Xie Z, Carrillo-Reid L, Auyeung G, Antonacci C, Buch A, Yang L, Beal MF, Surmeier DJ, Kordower JH, Tabar V, Studer L. Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson’s disease. Nature. 2011;480:547–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kikuchi T, Morizane A, Doi D, Magotani H, Onoe H, Hayashi T, Mizuma H, Takara S, Takahashi R, Inoue H, Morita S, Yamamoto M, Okita K, Nakagawa M, Parmar M, Takahashi J. Human iPS cell-derived dopaminergic neurons function in a primate Parkinson’s disease model. Nature. 2017;548:592–6.

    Article  CAS  PubMed  Google Scholar 

  77. Zheng X, Han D, Liu W, Wang X, Pan N, Wang Y, Chen Z. Human iPSC-derived midbrain organoids functionally integrate into striatum circuits and restore motor function in a mouse model of Parkinson’s disease. Theranostics. 2023;13:2673–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Grealish S, Diguet E, Kirkeby A, Mattsson B, Heuer A, Bramoulle Y, Van Camp N, Perrier AL, Hantraye P, Björklund A, Parmar M. Human ESC-derived dopamine neurons show similar preclinical efficacy and potency to fetal neurons when grafted in a rat model of Parkinson’s disease. Cell Stem Cell. 2014;15:653–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Doi D, Magotani H, Kikuchi T, Ikeda M, Hiramatsu S, Yoshida K, Amano N, Nomura M, Umekage M, Morizane A, Takahashi J. Pre-clinical study of induced pluripotent stem cell-derived dopaminergic progenitor cells for Parkinson’s disease. Nat Commun. 2020;11:3369.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Kim MS, Ra EA, Kweon SH, Seo BA, Ko HS, Oh Y, Lee G. Advanced human iPSC-based preclinical model for Parkinson’s disease with optogenetic alpha-synuclein aggregation. Cell Stem Cell. 2023;30:973–e98611.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Prudon N, Cordero-Espinoza L, Abarkan M, Gurchenkov B, Morel C, Lepleux M, De Luca V, Lartigue M, Cabanas H, Pujol N, Milvoy L, Morand P, Moncaubeig F, Wurtz H, Poinçot L, De Marco M, Jonckeau A, Pletenka J, Luquet E, Sovera A, Hardoüin J, Neves IJ, Machado-Hitau A, Schmit K, Piouceau L, Guilbert S, Manache-Alberici L, Lanero Fidalgo M, Dabée G, Dufourd T, Schroeder J, Alessandri K, Bezard E, Faggiani E, Feyeux M. Bioreactor-produced iPSCs-derived dopaminergic neuron-containing neural microtissues innervate and normalize rotational bias in a dose-dependent manner in a Parkinson rat model. Neurotherapeutics. 2024;21:e00436.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Song B, Cha Y, Ko S, Jeon J, Lee N, Seo H, Park KJ, Lee IH, Lopes C, Feitosa M, Luna MJ, Jung JH, Kim J, Hwang D, Cohen BM, Teicher MH, Leblanc P, Carter BS, Kordower JH, Bolshakov VY, Kong SW. Schweitzer JS and Kim KS. Human autologous iPSC-derived dopaminergic progenitors restore motor function in Parkinson’s disease models. J Clin Invest. 2020;130:904–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Takahashi J. Preclinical evaluation of patient-derived cells shows promise for Parkinson’s disease. J Clin Invest. 2020;130:601–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Piao J, Zabierowski S, Dubose BN, Hill EJ, Navare M, Claros N, Rosen S, Ramnarine K, Horn C, Fredrickson C, Wong K, Safford B, Kriks S, El Maarouf A, Rutishauser U, Henchcliffe C, Wang Y, Riviere I, Mann S, Bermudez V, Irion S, Studer L. Tomishima M and Tabar V. Preclinical efficacy and safety of a human embryonic stem Cell-Derived midbrain dopamine progenitor product, MSK-DA01. Cell Stem Cell. 2021;28:217–e2297.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Kirkeby A, Nelander J, Hoban DB, Rogelius N, Bjartmarz H, Storm P, Fiorenzano A, Adler AF, Vale S, Mudannayake J, Zhang Y, Cardoso T, Mattsson B, Landau AM, Glud AN, Sørensen JC, Lillethorup TP, Lowdell M, Carvalho C, Bain O, van Vliet T, Lindvall O, Björklund A, Harry B, Cutting E, Widner H, Paul G, Barker RA, Parmar M. 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–e13149.

    Article  CAS  PubMed  Google Scholar 

  86. Takahashi J. Strategies for bringing stem cell-derived dopamine neurons to the clinic: the Kyoto trial. Prog Brain Res. 2017;230:213–26.

    Article  PubMed  Google Scholar 

  87. Barker RA, Parmar M, Studer L, Takahashi J. Human trials of stem Cell-Derived dopamine neurons for Parkinson’s disease: dawn of a new era. Cell Stem Cell. 2017;21:569–73.

    Article  CAS  PubMed  Google Scholar 

  88. Schweitzer JS, Song B, Herrington TM, Park TY, Lee N, Ko S, Jeon J, Cha Y, Kim K, Li Q, Henchcliffe C, Kaplitt M, Neff C, Rapalino O, Seo H, Lee IH, Kim J, Kim T, Petsko GA, Ritz J, Cohen BM, Kong SW, Leblanc P. Carter BS and Kim KS. Personalized iPSC-Derived dopamine progenitor cells for Parkinson’s disease. N Engl J Med. 2020;382:1926–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Park S, Park CW, Eom JH, Jo MY, Hur HJ, Choi SK, Lee JS, Nam ST, Jo KS, Oh YW, Lee J, Kim S, Kim DH, Park CY, Kim SJ, Lee HY, Cho MS, Kim DS, Kim DW. Preclinical and dose-ranging assessment of hESC-derived dopaminergic progenitors for a clinical trial on Parkinson’s disease. Cell Stem Cell. 2024;31:25–e388.

    Article  CAS  PubMed  Google Scholar 

  90. Clark BJ, Lelos MJ, Loring JF. Advancing Parkinson’s disease treatment: cell replacement therapy with neurons derived from pluripotent stem cells. Stem Cells. 2024;42:781–90.

    Article  PubMed  Google Scholar 

  91. Yang J, Li S, He XB, Cheng C, Le W. Induced pluripotent stem cells in Alzheimer’s disease: applications for disease modeling and cell-replacement therapy. Mol Neurodegener. 2016;11:39.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Espuny-Camacho I, Arranz AM, Fiers M, Snellinx A, Ando K, Munck S, Bonnefont J, Lambot L, Corthout N, Omodho L, Vanden Eynden E, Radaelli E, Tesseur I, Wray S, Ebneth A, Hardy J, Leroy K, Brion JP, Vanderhaeghen P, De Strooper B. Hallmarks of Alzheimer’s disease in Stem-Cell-Derived human neurons transplanted into mouse brain. Neuron. 2017;93:1066–e10818.

    Article  CAS  PubMed  Google Scholar 

  93. Zhu Q, Zhang N, Hu N, Jiang R, Lu H, Xuan A, Long D, Chen Y. Neural stem cell transplantation improves learning and memory by protecting cholinergic neurons and restoring synaptic impairment in an amyloid precursor protein/presenilin 1 Transgenic mouse model of Alzheimer’s disease. Mol Med Rep. 2020;21:1172–80.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Penney J, Ralvenius WT, Tsai LH. Modeling Alzheimer’s disease with iPSC-derived brain cells. Mol Psychiatry. 2020;25:148–67.

    Article  PubMed  Google Scholar 

  95. Wang ZB, Wang ZT, Sun Y, Tan L, Yu JT. The future of stem cell therapies of Alzheimer’s disease. Ageing Res Rev. 2022;80:101655.

    Article  CAS  PubMed  Google Scholar 

  96. Yagi T, Ito D, Okada Y, Akamatsu W, Nihei Y, Yoshizaki T, Yamanaka S, Okano H, Suzuki N. Modeling Familial Alzheimer’s disease with induced pluripotent stem cells. Hum Mol Genet. 2011;20:4530–9.

    Article  CAS  PubMed  Google Scholar 

  97. Israel MA, Yuan SH, Bardy C, Reyna SM, Mu Y, Herrera C, Hefferan MP, Van Gorp S, Nazor KL, Boscolo FS, Carson CT, Laurent LC, Marsala M, Gage FH, Remes AM, Koo EH, Goldstein LS. Probing sporadic and Familial Alzheimer’s disease using induced pluripotent stem cells. Nature. 2012;482:216–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Fujiwara N, Shimizu J, Takai K, Arimitsu N, Saito A, Kono T, Umehara T, Ueda Y, Wakisaka S, Suzuki T, Suzuki N. Restoration of Spatial memory dysfunction of human APP Transgenic mice by transplantation of neuronal precursors derived from human iPS cells. Neurosci Lett. 2013;557 Pt B:129–34.

    Article  PubMed  Google Scholar 

  99. Tao R, Lu R, Wang J, Zeng S, Zhang T, Guo W, Zhang X, Cheng Q, Yue C, Wang Y, Jing N. Probing the therapeutic potential of TRPC6 for Alzheimer’s disease in live neurons from patient-specific iPSCs. J Mol Cell Biol. 2020;12:807–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Tao R, Yue C, Guo Z, Guo W, Yao Y, Yang X, Shao Z, Gao C, Ding J, Shen L, Chen S, Jing N. Subtype-specific neurons from patient iPSCs display distinct neuropathological features of Alzheimer’s disease. Cell Regen. 2024;13:21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Burkhardt MF, Martinez FJ, Wright S, Ramos C, Volfson D, Mason M, Garnes J, Dang V, Lievers J, Shoukat-Mumtaz U, Martinez R, Gai H, Blake R, Vaisberg E, Grskovic M, Johnson C, Irion S, Bright J, Cooper B, Nguyen L. Griswold-Prenner I and Javaherian A. A cellular model for sporadic ALS using patient-derived induced pluripotent stem cells. Mol Cell Neurosci. 2013;56:355–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Lee JH, Liu JW, Lin SZ, Harn HJ, Chiou TW. Advances in Patient-Specific induced pluripotent stem cells shed light on drug discovery for amyotrophic lateral sclerosis. Cell Transpl. 2018;27:1301–12.

    Article  Google Scholar 

  103. Hawrot J, Imhof S, Wainger BJ. Modeling cell-autonomous motor neuron phenotypes in ALS using iPSCs. Neurobiol Dis. 2020;134:104680.

    Article  CAS  PubMed  Google Scholar 

  104. Giacomelli E, Vahsen BF, Calder EL, Xu Y, Scaber J, Gray E, Dafinca R, Talbot K, Studer L. Human stem cell models of neurodegeneration: from basic science of amyotrophic lateral sclerosis to clinical translation. Cell Stem Cell. 2022;29:11–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Kondo T, Funayama M, Tsukita K, Hotta A, Yasuda A, Nori S, Kaneko S, Nakamura M, Takahashi R, Okano H, Yamanaka S, Inoue H. Focal transplantation of human iPSC-derived glial-rich neural progenitors improves lifespan of ALS mice. Stem Cell Rep. 2014;3:242–9.

    Article  CAS  Google Scholar 

  106. Nizzardo M, Simone C, Rizzo F, Ruggieri M, Salani S, Riboldi G, Faravelli I, Zanetta C, Bresolin N, Comi GP, Corti S. Minimally invasive transplantation of iPSC-derived ALDHhiSSCloVLA4 + neural stem cells effectively improves the phenotype of an amyotrophic lateral sclerosis model. Hum Mol Genet. 2014;23:342–54.

    Article  CAS  PubMed  Google Scholar 

  107. Popescu IR, Nicaise C, Liu S, Bisch G, Knippenberg S, Daubie V, Bohl D, Pochet R. Neural progenitors derived from human induced pluripotent stem cells survive and differentiate upon transplantation into a rat model of amyotrophic lateral sclerosis. Stem Cells Transl Med. 2013;2:167–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Liu B, Li M, Zhang L, Chen Z, Lu P. Motor neuron replacement therapy for amyotrophic lateral sclerosis. Neural Regen Res. 2022;17:1633–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Brix J, Zhou Y, Luo Y. The epigenetic reprogramming roadmap in generation of iPSCs from somatic cells. J Genet Genomics. 2015;42:661–70.

    Article  CAS  PubMed  Google Scholar 

  110. Sato Y, Bando H, Di Piazza M, Gowing G, Herberts C, Jackman S, Leoni G, Libertini S, MacLachlan T, McBlane JW, Pereira Mouriès L, Sharpe M, Shingleton W, Surmacz-Cordle B, Yamamoto K, van der Laan JW. Tumorigenicity assessment of cell therapy products: the need for global consensus and points to consider. Cytotherapy. 2019;21:1095–111.

    Article  CAS  PubMed  Google Scholar 

  111. Bogomiakova ME, Sekretova EK, Anufrieva KS, Khabarova PO, Kazakova AN, Bobrovsky PA, Grigoryeva TV, Eremeev AV, Lebedeva OS, Bogomazova AN, Lagarkova MA. iPSC-derived cells lack immune tolerance to autologous NK-cells due to imbalance in ligands for activating and inhibitory NK-cell receptors. Stem Cell Res Ther. 2023;14:77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Petrus-Reurer S, Romano M, Howlett S, Jones JL, Lombardi G, Saeb-Parsy K. Immunological considerations and challenges for regenerative cellular therapies. Commun Biol. 2021;4:798.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Jin H, Zhang YT, Yang Y, Wen LY, Wang JH, Xu HY, Lai BQ, Feng B, Che MT, Qiu XC, Li ZL, Wang LJ, Ruan JW, Jiang B, Zeng X, Deng QW, Li G, Ding Y, Zeng YS. Electroacupuncture facilitates the integration of neural stem Cell-Derived neural network with transected rat spinal cord. Stem Cell Rep. 2019;12:274–89.

    Article  Google Scholar 

  114. Patil N, Korenfeld O, Scalf RN, Lavoie N, Huntemer-Silveira A, Han G, Swenson R, Parr AM. Electrical stimulation affects the differentiation of transplanted regionally specific human spinal neural progenitor cells (sNPCs) after chronic spinal cord injury. Stem Cell Res Ther. 2023;14:378.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. da Silva Meirelles L, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci. 2006;119:2204–13.

    Article  PubMed  Google Scholar 

  116. Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet. 1970;3:393–403.

    CAS  PubMed  Google Scholar 

  117. Rodriguez AM, Elabd C, Amri EZ, Ailhaud G, Dani C. The human adipose tissue is a source of multipotent stem cells. Biochimie. 2005;87:125–8.

    Article  CAS  PubMed  Google Scholar 

  118. Zhang X, Hirai M, Cantero S, Ciubotariu R, Dobrila L, Hirsh A, Igura K, Satoh H, Yokomi I, Nishimura T, Yamaguchi S, Yoshimura K, Rubinstein P, Takahashi TA. Isolation and characterization of mesenchymal stem cells from human umbilical cord blood: Reevaluation of critical factors for successful isolation and high ability to proliferate and differentiate to chondrocytes as compared to mesenchymal stem cells from bone marrow and adipose tissue. J Cell Biochem. 2011;112:1206–18.

    Article  CAS  PubMed  Google Scholar 

  119. Samsonraj RM, Raghunath M, Nurcombe V, Hui JH, van Wijnen AJ, Cool SM. Concise review: multifaceted characterization of human mesenchymal stem cells for use in regenerative medicine. Stem Cells Transl Med. 2017;6:2173–85.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Aliaghaei A, Boroujeni ME, Ahmadi H, Bayat AH, Tavirani MR, Abdollahifar MA, Pooyafar MH, Mansouri V. Dental pulp stem cell transplantation ameliorates motor function and prevents cerebellar atrophy in rat model of cerebellar ataxia. Cell Tissue Res. 2019;376:179–87.

    Article  PubMed  Google Scholar 

  121. Marion NW, Mao JJ. Mesenchymal stem cells and tissue engineering. Methods Enzymol. 2006;420:339–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Fraile M, Eiro N, Costa LA, Martín A, Vizoso FJ. Aging and mesenchymal stem cells: basic concepts, challenges and strategies. Biology (Basel). 2022;11.

  123. Zhu Y, Ge J, Huang C, Liu H, Jiang H. Application of mesenchymal stem cell therapy for aging frailty: from mechanisms to therapeutics. Theranostics. 2021;11:5675–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Le Blanc K, Ringdén O. Immunomodulation by mesenchymal stem cells and clinical experience. J Intern Med. 2007;262:509–25.

    Article  PubMed  Google Scholar 

  125. Khan MI, Jeong ES, Khan MZ, Shin JH, Kim JD. Stem cells-derived exosomes alleviate neurodegeneration and Alzheimer’s pathogenesis by ameliorating neuroinflamation, and regulating the associated molecular pathways. Sci Rep. 2023;13:15731.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Lo Furno D, Mannino G, Giuffrida R. Functional role of mesenchymal stem cells in the treatment of chronic neurodegenerative diseases. J Cell Physiol. 2018;233:3982–99.

    Article  CAS  PubMed  Google Scholar 

  127. Duncan T, Valenzuela M. Alzheimer’s disease, dementia, and stem cell therapy. Stem Cell Res Ther. 2017;8:111.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Doshmanziari M, Shirian S, Kouchakian MR, Moniri SF, Jangnoo S, Mohammadi N, Zafari F. Mesenchymal stem cells act as stimulators of neurogenesis and synaptic function in a rat model of Alzheimer’s disease. Heliyon. 2021;7:e07996.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Hernández AE, García E. Mesenchymal stem cell therapy for Alzheimer’s disease. Stem Cells Int. 2021;2021:7834421.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Shin JY, Park HJ, Kim HN, Oh SH, Bae JS, Ha HJ, Lee PH. Mesenchymal stem cells enhance autophagy and increase β-amyloid clearance in alzheimer disease models. Autophagy. 2014;10:32–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Santamaria G, Brandi E, Vitola P, Grandi F, Ferrara G, Pischiutta F, Vegliante G, Zanier ER, Re F, Uccelli A, Forloni G, de Rosbo NK, Balducci C. Intranasal delivery of mesenchymal stem cell secretome repairs the brain of Alzheimer’s mice. Cell Death Differ. 2021;28:203–18.

    Article  CAS  PubMed  Google Scholar 

  132. Zheng XY, Wan QQ, Zheng CY, Zhou HL, Dong XY, Deng QS, Yao H, Fu Q, Gao M, Yan ZJ, Wang SS, You Y, Lv J, Wang XY, Chen KE, Zhang MY, Xu RX. Amniotic mesenchymal stem cells decrease Aβ deposition and improve memory in APP/PS1 Transgenic mice. Neurochem Res. 2017;42:2191–207.

    Article  CAS  PubMed  Google Scholar 

  133. Kim HJ, Seo SW, Chang JW, Lee JI, Kim CH, Chin J, Choi SJ, Kwon H, Yun HJ, Lee JM, Kim ST, Choe YS, Lee KH, Na DL. Stereotactic brain injection of human umbilical cord blood mesenchymal stem cells in patients with Alzheimer’s disease dementia: A phase 1 clinical trial. Alzheimers Dement (N Y). 2015;1:95–102.

    Article  PubMed  Google Scholar 

  134. Kim HJ, Cho KR, Jang H, Lee NK, Jung YH, Kim JP, Lee JI, Chang JW, Park S, Kim ST, Moon SW, Seo SW, Choi SJ, Na DL. Intracerebroventricular injection of human umbilical cord blood mesenchymal stem cells in patients with Alzheimer’s disease dementia: a phase I clinical trial. Alzheimers Res Ther. 2021;13:154.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Danielyan L, Beer-Hammer S, Stolzing A, Schäfer R, Siegel G, Fabian C, Kahle P, Biedermann T, Lourhmati A, Buadze M, Novakovic A, Proksch B, Gleiter CH, Frey WH, Schwab M. Intranasal delivery of bone marrow-derived mesenchymal stem cells, macrophages, and microglia to the brain in mouse models of Alzheimer’s and Parkinson’s disease. Cell Transpl. 2014;23(Suppl 1):S123–39.

    Article  Google Scholar 

  136. Venkataramana NK, Kumar SK, Balaraju S, Radhakrishnan RC, Bansal A, Dixit A, Rao DK, Das M, Jan M, Gupta PK, Totey SM. Open-labeled study of unilateral autologous bone-marrow-derived mesenchymal stem cell transplantation in Parkinson’s disease. Transl Res. 2010;155:62–70.

    Article  CAS  PubMed  Google Scholar 

  137. Boika A, Aleinikava N, Chyzhyk V, Zafranskaya M, Nizheharodava D, Ponomarev V. Mesenchymal stem cells in Parkinson’s disease: motor and nonmotor symptoms in the early posttransplant period. Surg Neurol Int. 2020;11:380.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Gavasso S, Kråkenes T, Olsen H, Evjenth EC, Ytterdal M, Haugsøen JB, Kvistad CE. The therapeutic mechanisms of mesenchymal stem cells in MS-A review focusing on neuroprotective properties. Int J Mol Sci. 2024;25.

  139. Kim SH, Oh KW, Noh MY, Kwon MS. Optimal therapeutic strategy of bone Marrow-Originated autologous mesenchymal stromal/stem cells for ALS. Stem Cells Transl Med. 2024;13:309–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Terashima T, Kobashi S, Watanabe Y, Nakanishi M, Honda N, Katagi M, Ohashi N, Kojima H. Enhancing the therapeutic efficacy of bone Marrow-Derived mononuclear cells with growth Factor-Expressing mesenchymal stem cells for ALS in mice. iScience. 2020;23:101764.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Harris VK, Yan QJ, Vyshkina T, Sahabi S, Liu X, Sadiq SA. Clinical and pathological effects of intrathecal injection of mesenchymal stem cell-derived neural progenitors in an experimental model of multiple sclerosis. J Neurol Sci. 2012;313:167–77.

    Article  PubMed  Google Scholar 

  142. Syková E, Rychmach P, Drahorádová I, Konrádová Š, Růžičková K, Voříšek I, Forostyak S, Homola A, Bojar M. Transplantation of mesenchymal stromal cells in patients with amyotrophic lateral sclerosis: results of phase I/IIa clinical trial. Cell Transpl. 2017;26:647–58.

    Article  Google Scholar 

  143. Berry JD, Cudkowicz ME, Windebank AJ, Staff NP, Owegi M, Nicholson K, McKenna-Yasek D, Levy YS, Abramov N, Kaspi H, Mehra M, Aricha R, Gothelf Y, Brown RH, NurOwn. Phase 2, randomized, clinical trial in patients with ALS: safety, clinical, and biomarker results. Neurology. 2019;93:e2294–305.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Cudkowicz ME, Lindborg SR, Goyal NA, Miller RG, Burford MJ, Berry JD, Nicholson KA, Mozaffar T, Katz JS, Jenkins LJ, Baloh RH, Lewis RA, Staff NP, Owegi MA, Berry DA, Gothelf Y, Levy YS, Aricha R, Kern RZ, Windebank AJ, Brown RH. Jr. A randomized placebo-controlled phase 3 study of mesenchymal stem cells induced to secrete high levels of neurotrophic factors in amyotrophic lateral sclerosis. Muscle Nerve. 2022;65:291–302.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Petrou P, Kassis I, Yaghmour NE, Ginzberg A, Karussis D. A phase II clinical trial with repeated intrathecal injections of autologous mesenchymal stem cells in patients with amyotrophic lateral sclerosis. Front Biosci (Landmark Ed). 2021;26:693–706.

    Article  CAS  PubMed  Google Scholar 

  146. Najar M, Melki R, Khalife F, Lagneaux L, Bouhtit F, Moussa Agha D, Fahmi H, Lewalle P, Fayyad-Kazan M, Merimi M. Therapeutic mesenchymal stem/stromal cells: value, challenges and optimization. Front Cell Dev Biol. 2021;9:716853.

    Article  PubMed  Google Scholar 

  147. Levy O, Kuai R, Siren EMJ, Bhere D, Milton Y, Nissar N, De Biasio M, Heinelt M, Reeve B, Abdi R, Alturki M, Fallatah M, Almalik A, Alhasan AH, Shah K, Karp JM. Shattering barriers toward clinically meaningful MSC therapies. Sci Adv. 2020;6:eaba6884.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Fernández-Santos ME, Garcia-Arranz M, Andreu EJ, García-Hernández AM, López-Parra M, Villarón E, Sepúlveda P, Fernández-Avilés F, García-Olmo D, Prosper F, Sánchez-Guijo F, Moraleda JM, Zapata AG. Optimization of mesenchymal stromal cell (MSC) manufacturing processes for a better therapeutic outcome. Front Immunol. 2022;13:918565.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Dunn CM, Kameishi S, Grainger DW, Okano T. Strategies to address mesenchymal stem/stromal cell heterogeneity in Immunomodulatory profiles to improve cell-based therapies. Acta Biomater. 2021;133:114–25.

    Article  CAS  PubMed  Google Scholar 

  150. Yang L, Liu SC, Liu YY, Zhu FQ, Xiong MJ, Hu DX, Zhang WJ. Therapeutic role of neural stem cells in neurological diseases. Front Bioeng Biotechnol. 2024;12:1329712.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Zhang J, Jiao J. Molecular biomarkers for embryonic and adult neural stem cell and neurogenesis. Biomed Res Int. 2015;2015:727542.

    PubMed  PubMed Central  Google Scholar 

  152. Galiakberova AA, Dashinimaev EB. Neural stem cells and methods for their generation from induced pluripotent stem cells in vitro. Front Cell Dev Biol. 2020;8:815.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Erharter A, Rizzi S, Mertens J, Edenhofer F. Take the shortcut - direct conversion of somatic cells into induced neural stem cells and their biomedical applications. FEBS Lett. 2019;593:3353–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Navarro Negredo P, Yeo RW, Brunet A. Aging and rejuvenation of neural stem cells and their niches. Cell Stem Cell. 2020;27:202–23.

    Article  CAS  PubMed  Google Scholar 

  155. Culig L, Chu X, Bohr VA. Neurogenesis in aging and age-related neurodegenerative diseases. Ageing Res Rev. 2022;78:101636.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Velikic G, Maric DM, Maric DL, Supic G, Puletic M, Dulic O, Vojvodic D. Harnessing the stem cell niche in regenerative medicine: innovative avenue to combat neurodegenerative diseases. Int J Mol Sci. 2024;25(2):993. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms25020993

  157. Ottoboni L, Merlini A, Martino G. Neural stem cell plasticity: advantages in therapy for the injured central nervous system. Front Cell Dev Biol. 2017;5:52.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Knippenberg S, Rath KJ, Böselt S, Thau-Habermann N, Schwarz SC, Dengler R, Wegner F, Petri S. Intraspinal administration of human spinal cord-derived neural progenitor cells in the G93A-SOD1 mouse model of ALS delays symptom progression, prolongs survival and increases expression of endogenous neurotrophic factors. J Tissue Eng Regen Med. 2017;11:751–64.

    Article  CAS  PubMed  Google Scholar 

  159. Teng YD, Benn SC, Kalkanis SN, Shefner JM, Onario RC, Cheng B, Lachyankar MB, Marconi M, Li J, Yu D, Han I, Maragakis NJ, Lládo J, Erkmen K, Redmond DE Jr., Sidman RL, Przedborski S, Rothstein JD, Brown RH. Jr. and snyder EY. Multimodal actions of neural stem cells in a mouse model of ALS: a meta-analysis. Sci Transl Med. 2012;4:165ra164.

    Article  PubMed  Google Scholar 

  160. Forostyak S, Forostyak O, Kwok JCF, Romanyuk N, Rehorova M, Kriska J, Dayanithi G, Raha-Chowdhury R, Jendelova P, Anderova M, Fawcett JW, Sykova E. Transplantation of neural precursors derived from induced pluripotent cells preserve perineuronal Nets and stimulate neural plasticity in ALS rats. Int J Mol Sci. 2020;21.

  161. Glass JD, Boulis NM, Johe K, Rutkove SB, Federici T, Polak M, Kelly C, Feldman EL. Lumbar intraspinal injection of neural stem cells in patients with amyotrophic lateral sclerosis: results of a phase I trial in 12 patients. Stem Cells. 2012;30:1144–51.

    Article  CAS  PubMed  Google Scholar 

  162. Feldman EL, Boulis NM, Hur J, Johe K, Rutkove SB, Federici T, Polak M, Bordeau J, Sakowski SA, Glass JD. Intraspinal neural stem cell transplantation in amyotrophic lateral sclerosis: phase 1 trial outcomes. Ann Neurol. 2014;75:363–73.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Riley J, Federici T, Polak M, Kelly C, Glass J, Raore B, Taub J, Kesner V, Feldman EL, Boulis NM. Intraspinal stem cell transplantation in amyotrophic lateral sclerosis: a phase I safety trial, technical note, and lumbar safety outcomes. Neurosurgery. 2012;71:405– 16; discussion 416.

  164. Mazzini L, Gelati M, Profico DC, Sgaravizzi G, Projetti Pensi M, Muzi G, Ricciolini C, Rota Nodari L, Carletti S, Giorgi C, Spera C, Domenico F, Bersano E, Petruzzelli F, Cisari C, Maglione A, Sarnelli MF, Stecco A, Querin G, Masiero S, Cantello R, Ferrari D, Zalfa C, Binda E, Visioli A, Trombetta D, Novelli A, Torres B, Bernardini L, Carriero A, Prandi P, Servo S, Cerino A, Cima V, Gaiani A, Nasuelli N, Massara M, Glass J, Sorarù G, Boulis NM, Vescovi AL. Human neural stem cell transplantation in ALS: initial results from a phase I trial. J Transl Med. 2015;13:17.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Baloh RH, Johnson JP, Avalos P, Allred P, Svendsen S, Gowing G, Roxas K, Wu A, Donahue B, Osborne S, Lawless G, Shelley B, Wheeler K, Prina C, Fine D, Kendra-Romito T, Stokes H, Manoukian V, Muthukumaran A, Garcia L, Bañuelos MG, Godoy M, Bresee C, Yu H, Drazin D, Ross L, Naruse R, Babu H, Macklin EA, Vo A, Elsayegh A, Tourtellotte W, Maya M, Burford M, Diaz F, Patil CG, Lewis RA, Svendsen CN. Transplantation of human neural progenitor cells secreting GDNF into the spinal cord of patients with ALS: a phase 1/2a trial. Nat Med. 2022;28:1813–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Leone MA, Gelati M, Profico DC, Gobbi C, Pravatà E, Copetti M, Conti C, Abate L, Amoruso L, Apollo F, Balzano RF, Bicchi I, Carella M, Ciampini A, Colosimo C, Crociani P, D’Aloisio G, Di Viesti P, Ferrari D, Fogli D, Fontana A, Frondizi D, Grespi V, Kuhle J, Laborante A, Lombardi I, Muzi G, Paci F, Placentino G, Popolizio T, Ricciolini C, Sabatini S, Silveri G, Spera C, Stephenson D, Stipa G, Tinella E, Zarrelli M, Zecca C, Ventura Y, D’Alessandro A, Peruzzotti-Jametti L, Pluchino S, Vescovi AL. Phase I clinical trial of intracerebroventricular transplantation of allogeneic neural stem cells in people with progressive multiple sclerosis. Cell Stem Cell. 2023;30:1597–e16098.

    Article  CAS  PubMed  Google Scholar 

  167. Mazzini L, Gelati M, Profico DC, Sorarù G, Ferrari D, Copetti M, Muzi G, Ricciolini C, Carletti S, Giorgi C, Spera C, Frondizi D, Masiero S, Stecco A, Cisari C, Bersano E, De Marchi F, Sarnelli MF, Querin G, Cantello R, Petruzzelli F, Maglione A, Zalfa C, Binda E, Visioli A, Trombetta D, Torres B, Bernardini L, Gaiani A, Massara M, Paolucci S, Boulis NM, Vescovi AL. Results from phase I clinical trial with intraspinal injection of neural stem cells in amyotrophic lateral sclerosis: A Long-Term outcome. Stem Cells Transl Med. 2019;8:887–97.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Glass JD, Hertzberg VS, Boulis NM, Riley J, Federici T, Polak M, Bordeau J, Fournier C, Johe K, Hazel T, Cudkowicz M, Atassi N, Borges LF, Rutkove SB, Duell J, Patil PG, Goutman SA, Feldman EL. Transplantation of spinal cord-derived neural stem cells for ALS: analysis of phase 1 and 2 trials. Neurology. 2016;87:392–400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Willis CM, Nicaise AM, Peruzzotti-Jametti L, Pluchino S. The neural stem cell secretome and its role in brain repair. Brain Res. 2020;1729:146615.

    Article  PubMed  Google Scholar 

  170. Lee EJ, Choi Y, Lee HJ, Hwang DW, Lee DS. Human neural stem cell-derived extracellular vesicles protect against Parkinson’s disease pathologies. J Nanobiotechnol. 2022;20:198.

    Article  CAS  Google Scholar 

  171. Ni W, Zhou J, Ling Y, Lu X, Niu D, Zeng Y, Qiu Y, Si Y, Wang J, Zhang W, Wang Z, Hu J. Neural stem cell secretome exerts a protective effect on damaged neuron mitochondria in Parkinson’s disease model. Brain Res. 2022;1790:147978.

    Article  CAS  PubMed  Google Scholar 

  172. Kim SW, Woo HJ, Kim EH, Kim HS, Suh HN, Kim SH, Song JJ, Wulansari N, Kang M, Choi SY, Choi SJ, Jang WH, Lee J, Kim KH, Lee W, Kim SH, Yang J, Kyung J, Lee HS, Park SM, Chang MY, Lee SH. Neural stem cells derived from human midbrain organoids as a stable source for treating Parkinson’s disease: midbrain organoid-NSCs (Og-NSC) as a stable source for PD treatment. Prog Neurobiol. 2021;204:102086.

    Article  CAS  PubMed  Google Scholar 

  173. Le Grand JN, Gonzalez-Cano L, Pavlou MA, Schwamborn JC. Neural stem cells in Parkinson’s disease: a role for neurogenesis defects in onset and progression. Cell Mol Life Sci. 2015;72:773–97.

    Article  PubMed  Google Scholar 

  174. Choi DH, Kim JH, Kim SM, Kang K, Han DW, Lee J. Therapeutic Potential of Induced Neural Stem Cells for Parkinson’s Disease. Int J Mol Sci. 2017;18.

  175. Nelke A, García-López S, Martínez-Serrano A, Pereira MP. Multifactoriality of Parkinson’s disease as explored through human neural stem cells and their transplantation in Middle-Aged parkinsonian mice. Front Pharmacol. 2021;12:773925.

    Article  CAS  PubMed  Google Scholar 

  176. Madrazo I, Kopyov O, Ávila-Rodríguez MA, Ostrosky F, Carrasco H, Kopyov A, Avendaño-Estrada A, Jiménez F, Magallón E, Zamorano C, González G, Valenzuela T, Carrillo R, Palma F, Rivera R. Franco-Bourland RE and Guízar-Sahagún G. Transplantation of human neural progenitor cells (NPC) into putamina of parkinsonian patients: A case series study, safety and efficacy four years after surgery. Cell Transpl. 2019;28:269–85.

    Article  CAS  Google Scholar 

  177. Chen Z, Zhao G. First-in-human transplantation of autologous induced neural stem cell-derived dopaminergic precursors to treat Parkinson’s disease. Sci Bull (Beijing). 2023;68:2700–3.

    Article  CAS  PubMed  Google Scholar 

  178. Lige L, Zengmin T. Transplantation of neural precursor cells in the treatment of Parkinson disease: an efficacy and safety analysis. Turk Neurosurg. 2016;26:378–83.

    PubMed  Google Scholar 

  179. Boese AC, Hamblin MH, Lee JP. Neural stem cell therapy for neurovascular injury in Alzheimer’s disease. Exp Neurol. 2020;324:113112.

    Article  CAS  PubMed  Google Scholar 

  180. Li X, Zhu H, Sun X, Zuo F, Lei J, Wang Z, Bao X, Wang R. Human neural stem cell transplantation rescues cognitive defects in APP/PS1 model of Alzheimer’s disease by enhancing neuronal connectivity and metabolic activity. Front Aging Neurosci. 2016;8:282.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Xuan AG, Long DH, Gu HG, Yang DD, Hong LP, Leng SL. BDNF improves the effects of neural stem cells on the rat model of Alzheimer’s disease with unilateral lesion of fimbria-fornix. Neurosci Lett. 2008;440:331–5.

    Article  CAS  PubMed  Google Scholar 

  182. Ebrahimi MJ, Aliaghaei A, Boroujeni ME, Khodagholi F, Meftahi G, Abdollahifar MA, Ahmadi H, Danyali S, Daftari M, Sadeghi Y. Human umbilical cord matrix stem cells reverse oxidative Stress-Induced cell death and ameliorate motor function and striatal atrophy in rat model of huntington disease. Neurotox Res. 2018;34:273–84.

    Article  CAS  PubMed  Google Scholar 

  183. Fan XL, Zhang Y, Li X, Fu QL. Mechanisms underlying the protective effects of mesenchymal stem cell-based therapy. Cell Mol Life Sci. 2020;77:2771–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Zhao L, Liu JW, Shi HY, Ma YM. Neural stem cell therapy for brain disease. World J Stem Cells. 2021;13:1278–92.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Chang J, Li Y, Shan X, Chen X, Yan X, Liu J, Zhao L. Neural stem cells promote neuroplasticity: a promising therapeutic strategy for the treatment of Alzheimer’s disease. Neural Regen Res. 2024;19:619–28.

    Article  CAS  PubMed  Google Scholar 

  186. Grade S, Götz M. Neuronal replacement therapy: previous achievements and challenges ahead. NPJ Regen Med. 2017;2:29.

    Article  PubMed  PubMed Central  Google Scholar 

  187. Götz M, Bocchi R. Neuronal replacement: concepts, achievements, and call for caution. Curr Opin Neurobiol. 2021;69:185–92.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Kirkeby A, Nolbrant S, Tiklova K, Heuer A, Kee N, Cardoso T, Ottosson DR, Lelos MJ, Rifes P, Dunnett SB, Grealish S, Perlmann T, Parmar M. Predictive markers guide differentiation to improve graft outcome in clinical translation of hESC-Based therapy for Parkinson’s disease. Cell Stem Cell. 2017;20:135–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Xiong M, Tao Y, Gao Q, Feng B, Yan W, Zhou Y, Kotsonis TA, Yuan T, You Z, Wu Z, Xi J, Haberman A, Graham J, Block J, Zhou W, Chen Y, Zhang SC. Human stem Cell-Derived neurons repair circuits and restore neural function. Cell Stem Cell. 2021;28:112–e1266.

    Article  CAS  PubMed  Google Scholar 

  190. Adler AF, Cardoso T, Nolbrant S, Mattsson B, Hoban DB, Jarl U, Wahlestedt JN, Grealish S. Björklund A and Parmar M. hESC-Derived dopaminergic transplants integrate into basal ganglia circuitry in a preclinical model of Parkinson’s disease. Cell Rep. 2019;28:3462–e34735.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Cardoso T, Adler AF, Mattsson B, Hoban DB, Nolbrant S, Wahlestedt JN, Kirkeby A, Grealish S, Björklund A, Parmar M. Target-specific forebrain projections and appropriate synaptic inputs of hESC-derived dopamine neurons grafted to the midbrain of parkinsonian rats. J Comp Neurol. 2018;526:2133–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Ma L, Hu B, Liu Y, Vermilyea SC, Liu H, Gao L, Sun Y, Zhang X, Zhang SC. Human embryonic stem cell-derived GABA neurons correct locomotion deficits in quinolinic acid-lesioned mice. Cell Stem Cell. 2012;10:455–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. 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;9:798–805.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Park H, Chang KA. Therapeutic Potential of Repeated Intravenous Transplantation of Human Adipose-Derived Stem Cells in Subchronic MPTP-Induced Parkinson’s Disease Mouse Model. Int J Mol Sci. 2020;21.

  195. Miranda M, Morici JF, Zanoni MB, Bekinschtein P. Brain-Derived neurotrophic factor: A key molecule for memory in the healthy and the pathological brain. Front Cell Neurosci. 2019;13:363.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Jia Y, Cao N, Zhai J, Zeng Q, Zheng P, Su R, Liao T, Liu J, Pei H, Fan Z, Zhou J, Xi J, He L, Chen L, Nan X, Yue W, Pei X. HGF mediates Clinical-Grade human umbilical Cord-Derived mesenchymal stem cells improved functional recovery in a Senescence-Accelerated mouse model of Alzheimer’s disease. Adv Sci (Weinh). 2020;7:1903809.

    Article  CAS  PubMed  Google Scholar 

  197. Kim DH, Lee D, Chang EH, Kim JH, Hwang JW, Kim JY, Kyung JW, Kim SH, Oh JS, Shim SM, Na DL, Oh W, Chang JW. GDF-15 secreted from human umbilical cord blood mesenchymal stem cells delivered through the cerebrospinal fluid promotes hippocampal neurogenesis and synaptic activity in an Alzheimer’s disease model. Stem Cells Dev. 2015;24:2378–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Xu R, Wu J, Lang L, Hu J, Tang H, Xu J, Sun B. Implantation of glial cell line-derived neurotrophic factor-expressing adipose tissue-derived stromal cells in a rat Parkinson’s disease model. Neurol Res. 2020;42:712–20.

    Article  CAS  PubMed  Google Scholar 

  199. Garcia KO, Ornellas FL, Martin PK, Patti CL, Mello LE, Frussa-Filho R, Han SW, Longo BM. Therapeutic effects of the transplantation of VEGF overexpressing bone marrow mesenchymal stem cells in the hippocampus of murine model of Alzheimer’s disease. Front Aging Neurosci. 2014;6:30.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Zhang P, Zhao G, Kang X, Su L. Effects of lateral ventricular transplantation of bone marrow-derived mesenchymal stem cells modified with brain-derived neurotrophic factor gene on cognition in a rat model of Alzheimer’s disease. Neural Regen Res. 2012;7:245–50.

    CAS  PubMed  PubMed Central  Google Scholar 

  201. Wang J, Hu W, Feng Z, Feng M. BDNF-overexpressing human umbilical cord mesenchymal stem cell-derived motor neurons improve motor function and prolong survival in amyotrophic lateral sclerosis mice. Neurol Res. 2021;43:199–209.

    Article  CAS  PubMed  Google Scholar 

  202. Pöyhönen S, Er S, Domanskyi A, Airavaara M. Effects of neurotrophic factors in glial cells in the central nervous system: expression and properties in neurodegeneration and injury. Front Physiol. 2019;10:486.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Li Y, Li F, Qin D, Chen H, Wang J, Wang J, Song S, Wang C, Wang Y, Liu S, Gao D, Wang ZH. The role of brain derived neurotrophic factor in central nervous system. Front Aging Neurosci. 2022;14:986443.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Schain M, Kreisl WC. Neuroinflammation in neurodegenerative Disorders-a review. Curr Neurol Neurosci Rep. 2017;17:25.

    Article  PubMed  Google Scholar 

  205. Kölliker-Frers R, Udovin L, Otero-Losada M, Kobiec T, Herrera MI, Palacios J, Razzitte G, Capani F. Neuroinflammation: an integrating overview of Reactive-Neuroimmune cell interactions in health and disease. Mediators Inflamm. 2021;2021:9999146.

    Article  PubMed  PubMed Central  Google Scholar 

  206. Zhang W, Xiao D, Mao Q, Xia H. Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther. 2023;8:267.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Li W, Ren G, Huang Y, Su J, Han Y, Li J, Chen X, Cao K, Chen Q, Shou P, Zhang L, Yuan ZR, Roberts AI, Shi S, Le AD, Shi Y. Mesenchymal stem cells: a double-edged sword in regulating immune responses. Cell Death Differ. 2012;19:1505–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Kook MG, Lee S, Shin N, Kong D, Kim DH, Kim MS, Kang HK, Choi SW, Kang KS. Repeated intramuscular transplantations of hUCB-MSCs improves motor function and survival in the SOD1 G(93)A mice through activation of AMPK. Sci Rep. 2020;10:1572.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Sironi F, Vallarola A, Violatto MB, Talamini L, Freschi M, De Gioia R, Capelli C, Agostini A, Moscatelli D, Tortarolo M, Bigini P, Introna M, Bendotti C. Multiple intracerebroventricular injections of human umbilical cord mesenchymal stem cells delay motor neurons loss but not disease progression of SOD1G93A mice. Stem Cell Res. 2017;25:166–78.

    Article  CAS  PubMed  Google Scholar 

  210. Zhang W, Sun HS, Wang X, Dumont AS, Liu Q. Cellular senescence, DNA damage, and neuroinflammation in the aging brain. Trends Neurosci. 2024;47:461–74.

    Article  CAS  PubMed  Google Scholar 

  211. Boukelmoune N, Chiu GS, Kavelaars A, Heijnen CJ. Mitochondrial transfer from mesenchymal stem cells to neural stem cells protects against the neurotoxic effects of cisplatin. Acta Neuropathol Commun. 2018;6:139.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Li H, Wang C, He T, Zhao T, Chen YY, Shen YL, Zhang X, Wang LL. Mitochondrial transfer from bone marrow mesenchymal stem cells to motor neurons in spinal cord injury rats via gap junction. Theranostics. 2019;9:2017–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Clemente-Suárez VJ, Martín-Rodríguez A, Yáñez-Sepúlveda R, Tornero-Aguilera JF. Mitochondrial transfer as a novel therapeutic approach in disease diagnosis and treatment. Int J Mol Sci. 2023;24.

  214. Beckervordersandforth R, Ebert B, Schäffner I, Moss J, Fiebig C, Shin J, Moore DL, Ghosh L, Trinchero MF, Stockburger C, Friedland K, Steib K, von Wittgenstein J, Keiner S, Redecker C, Hölter SM, Xiang W, Wurst W, Jagasia R, Schinder AF, Ming GL, Toni N, Jessberger S, Song H, Lie DC. Role of mitochondrial metabolism in the control of early lineage progression and aging phenotypes in adult hippocampal neurogenesis. Neuron. 2017;93:1518.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Paliwal S, Chaudhuri R, Agrawal A, Mohanty S. Regenerative abilities of mesenchymal stem cells through mitochondrial transfer. J Biomed Sci. 2018;25:31.

    Article  PubMed  PubMed Central  Google Scholar 

  216. Tan YL, Eng SP, Hafez P, Abdul Karim N, Law JX, Ng MH. Mesenchymal stromal cell mitochondrial transfer as a cell rescue strategy in regenerative medicine: A review of evidence in preclinical models. Stem Cells Transl Med. 2022;11:814–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Branco A, Moniz I, Ramalho-Santos J. Mitochondria as biological targets for stem cell and organismal senescence. Eur J Cell Biol. 2023;102:151289.

    Article  CAS  PubMed  Google Scholar 

  218. Choudhury D, Rong N, Senthil Kumar HV, Swedick S, Samuel RZ, Mehrotra P, Toftegaard J, Rajabian N, Thiyagarajan R, Podder AK, Wu Y, Shahini S, Seldeen KL, Troen B. Lei P and Andreadis ST. Proline restores mitochondrial function and reverses aging hallmarks in senescent cells. Cell Rep. 2024;43:113738.

    Article  CAS  PubMed  Google Scholar 

  219. Wang H, Yang Y, Liu J, Qian L. Direct cell reprogramming: approaches, mechanisms and progress. Nat Rev Mol Cell Biol. 2021;22:410–24.

    Article  PubMed  PubMed Central  Google Scholar 

  220. Xu J, Du Y, Deng H. Direct lineage reprogramming: strategies, mechanisms, and applications. Cell Stem Cell. 2015;16:119–34.

    Article  CAS  PubMed  Google Scholar 

  221. Bajohr J, Faiz M. Direct lineage reprogramming in the CNS. Adv Exp Med Biol. 2020;1212:31–48.

    Article  CAS  PubMed  Google Scholar 

  222. Bocchi R, Masserdotti G, Götz M. Direct neuronal reprogramming: fast forward from new concepts toward therapeutic approaches. Neuron. 2022;110:366–93.

    Article  CAS  PubMed  Google Scholar 

  223. Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Südhof TC, Wernig M. Direct conversion of fibroblasts to functional neurons by defined factors. Nature. 2010;463:1035–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Marro S, Pang ZP, Yang N, Tsai MC, Qu K, Chang HY, Südhof TC, Wernig M. Direct lineage conversion of terminally differentiated hepatocytes to functional neurons. Cell Stem Cell. 2011;9:374–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Kim J, Su SC, Wang H, Cheng AW, Cassady JP, Lodato MA, Lengner CJ, Chung CY, Dawlaty MM, Tsai LH, Jaenisch R. Functional integration of dopaminergic neurons directly converted from mouse fibroblasts. Cell Stem Cell. 2011;9:413–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Caiazzo M, Dell’Anno MT, Dvoretskova E, Lazarevic D, Taverna S, Leo D, Sotnikova TD, Menegon A, Roncaglia P, Colciago G, Russo G, Carninci P, Pezzoli G, Gainetdinov RR, Gustincich S. Dityatev A and broccoli V. Direct generation of functional dopaminergic neurons from mouse and human fibroblasts. Nature. 2011;476:224–7.

    Article  CAS  PubMed  Google Scholar 

  227. Ring KL, Tong LM, Balestra ME, Javier R, Andrews-Zwilling Y, Li G, Walker D, Zhang WR, Kreitzer AC, Huang Y. Direct reprogramming of mouse and human fibroblasts into multipotent neural stem cells with a single factor. Cell Stem Cell. 2012;11:100–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Sheng C, Zheng Q, Wu J, Xu Z, Wang L, Li W, Zhang H, Zhao XY, Liu L, Wang Z, Guo C, Wu HJ, Liu Z, Wang L, He S, Wang XJ, Chen Z, Zhou Q. Direct reprogramming of Sertoli cells into multipotent neural stem cells by defined factors. Cell Res. 2012;22:208–18.

    Article  CAS  PubMed  Google Scholar 

  229. Li X, Zuo X, Jing J, Ma Y, Wang J, Liu D, Zhu J, Du X, Xiong L, Du Y, Xu J, Xiao X, Wang J, Chai Z, Zhao Y, Deng H. Small-Molecule-Driven direct reprogramming of mouse fibroblasts into functional neurons. Cell Stem Cell. 2015;17:195–203.

    Article  CAS  PubMed  Google Scholar 

  230. Cheng L, Hu W, Qiu B, Zhao J, Yu Y, Guan W, Wang M, Yang W, Pei G. Generation of neural progenitor cells by chemical cocktails and hypoxia. Cell Res. 2014;24:665–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Zhang M, Lin YH, Sun YJ, Zhu S, Zheng J, Liu K, Cao N, Li K, Huang Y, Ding S. Pharmacological reprogramming of fibroblasts into neural stem cells by Signaling-Directed transcriptional activation. Cell Stem Cell. 2016;18:653–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Hu W, Qiu B, Guan W, Wang Q, Wang M, Li W, Gao L, Shen L, Huang Y, Xie G, Zhao H, Jin Y, Tang B, Yu Y, Zhao J, Pei G. Direct conversion of normal and Alzheimer’s disease human fibroblasts into neuronal cells by small molecules. Cell Stem Cell. 2015;17:204–12.

    Article  CAS  PubMed  Google Scholar 

  233. Zhang L, Yin JC, Yeh H, Ma NX, Lee G, Chen XA, Wang Y, Lin L, Chen L, Jin P, Wu GY, Chen G. Small molecules efficiently reprogram human astroglial cells into functional neurons. Cell Stem Cell. 2015;17:735–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Yin JC, Zhang L, Ma NX, Wang Y, Lee G, Hou XY, Lei ZF, Zhang FY, Dong FP, Wu GY, Chen G. Chemical conversion of human fetal astrocytes into neurons through modulation of multiple signaling pathways. Stem Cell Rep. 2019;12:488–501.

    Article  CAS  Google Scholar 

  235. Ma Y, Xie H, Du X, Wang L, Jin X, Zhang Q, Han Y, Sun S, Wang L, Li X, Zhang C, Wang M, Li C, Xu J, Huang Z, Wang X, Chai Z, Deng H. In vivo chemical reprogramming of astrocytes into neurons. Cell Discov. 2021;7:12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Pfisterer U, Kirkeby A, Torper O, Wood J, Nelander J, Dufour A, Björklund A, Lindvall O, Jakobsson J, Parmar M. Direct conversion of human fibroblasts to dopaminergic neurons. Proc Natl Acad Sci U S A. 2011;108:10343–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Liu X, Li F, Stubblefield EA, Blanchard B, Richards TL, Larson GA, He Y, Huang Q, Tan AC, Zhang D, Benke TA, Sladek JR, Zahniser NR, Li CY. Direct reprogramming of human fibroblasts into dopaminergic neuron-like cells. Cell Res. 2012;22:321–32.

    Article  CAS  PubMed  Google Scholar 

  238. Pang ZP, Yang N, Vierbuchen T, Ostermeier A, Fuentes DR, Yang TQ, Citri A, Sebastiano V, Marro S, Südhof TC, Wernig M. Induction of human neuronal cells by defined transcription factors. Nature. 2011;476:220–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Ambasudhan R, Talantova M, Coleman R, Yuan X, Zhu S, Lipton SA, Ding S. Direct reprogramming of adult human fibroblasts to functional neurons under defined conditions. Cell Stem Cell. 2011;9:113–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Yoo AS, Sun AX, Li L, Shcheglovitov A, Portmann T, Li Y, Lee-Messer C, Dolmetsch RE, Tsien RW, Crabtree GR. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature. 2011;476:228–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Son EY, Ichida JK, Wainger BJ, Toma JS, Rafuse VF, Woolf CJ, Eggan K. Conversion of mouse and human fibroblasts into functional spinal motor neurons. Cell Stem Cell. 2011;9:205–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Ofenbauer A, Tursun B. Strategies for in vivo reprogramming. Curr Opin Cell Biol. 2019;61:9–15.

    Article  CAS  PubMed  Google Scholar 

  243. Srivastava D, DeWitt N. In vivo cellular reprogramming: the next generation. Cell. 2016;166:1386–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Torper O, Pfisterer U, Wolf DA, Pereira M, Lau S, Jakobsson J, Björklund A, Grealish S, Parmar M. Generation of induced neurons via direct conversion in vivo. Proc Natl Acad Sci U S A. 2013;110:7038–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Guo Z, Zhang L, Wu Z, Chen Y, Wang F, Chen G. In vivo direct reprogramming of reactive glial cells into functional neurons after brain injury and in an Alzheimer’s disease model. Cell Stem Cell. 2014;14:188–202.

    Article  CAS  PubMed  Google Scholar 

  246. Wang J, Sun S, Deng H. Chemical reprogramming for cell fate manipulation: methods, applications, and perspectives. Cell Stem Cell. 2023;30:1130–47.

    Article  CAS  PubMed  Google Scholar 

  247. Li H, Chen G. In vivo reprogramming for CNS repair: regenerating neurons from endogenous glial cells. Neuron. 2016;91:728–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. Pereira M, Birtele M, Rylander Ottosson D. Direct reprogramming into interneurons: potential for brain repair. Cell Mol Life Sci. 2019;76:3953–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Ocampo A, Reddy P, Martinez-Redondo P, Platero-Luengo A, Hatanaka F, Hishida T, Li M, Lam D, Kurita M, Beyret E, Araoka T, Vazquez-Ferrer E, Donoso D, Roman JL, Xu J, Rodriguez Esteban C, Nuñez G, Nuñez Delicado E, Campistol JM, Guillen I, Guillen P, Izpisua Belmonte JC. In vivo amelioration of Age-Associated hallmarks by partial reprogramming. Cell. 2016;167:1719–e173312.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Singh PB, Zhakupova A. Age reprogramming: cell rejuvenation by partial reprogramming. Development. 2022;149.

  251. Gill D, Parry A, Santos F, Okkenhaug H, Todd CD, Hernando-Herraez I, Stubbs TM, Milagre I, Reik W. Multi-omic rejuvenation of human cells by maturation phase transient reprogramming. Elife. 2022;11.

  252. Sarkar TJ, Quarta M, Mukherjee S, Colville A, Paine P, Doan L, Tran CM, Chu CR, Horvath S, Qi LS, Bhutani N. Rando TA and Sebastiano V. Transient non-integrative expression of nuclear reprogramming factors promotes multifaceted amelioration of aging in human cells. Nat Commun. 2020;11:1545.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Browder KC, Reddy P, Yamamoto M, Haghani A, Guillen IG, Sahu S, Wang C, Luque Y, Prieto J, Shi L, Shojima K, Hishida T, Lai Z, Li Q, Choudhury FK, Wong WR, Liang Y, Sangaraju D, Sandoval W, Esteban CR, Delicado EN, Garcia PG, Pawlak M, Vander Heiden JA, Horvath S, Jasper H and Izpisua, Belmonte JC. In vivo partial reprogramming alters age-associated molecular changes during physiological aging in mice. Nat Aging. 2022;2:243–253.

  254. Wang C, Rabadan Ros R, Martinez-Redondo P, Ma Z, Shi L, Xue Y, Guillen-Guillen I, Huang L, Hishida T, Liao HK, Nuñez Delicado E, Rodriguez Esteban C, Guillen-Garcia P, Reddy P, Izpisua Belmonte JC. In vivo partial reprogramming of myofibers promotes muscle regeneration by remodeling the stem cell niche. Nat Commun. 2021;12:3094.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  255. Roux AE, Zhang C, Paw J, Zavala-Solorio J, Malahias E, Vijay T, Kolumam G, Kenyon C, Kimmel JC. Diverse partial reprogramming strategies restore youthful gene expression and transiently suppress cell identity. Cell Syst. 2022;13:574–e58711.

    Article  CAS  PubMed  Google Scholar 

  256. Yücel AD, Gladyshev VN. The long and winding road of reprogramming-induced rejuvenation. Nat Commun. 2024;15:1941.

    Article  PubMed  PubMed Central  Google Scholar 

  257. Macip CC, Hasan R, Hoznek V, Kim J, Lu YR, Metzger LEt, Sethna S, Davidsohn N. Gene Therapy-Mediated partial reprogramming extends lifespan and reverses Age-Related changes in aged mice. Cell Reprogram. 2024;26:24–32.

    Article  PubMed  PubMed Central  Google Scholar 

  258. Cho HE, Lee S, Seo JH, Kang SW, Choi WA, Cho SR. In vivo reprogramming using Yamanaka factors in the CNS: A scoping review. Cells. 2024;13.

  259. Sichani AS, Khoddam S, Shakeri S, Tavakkoli Z, Jafroodi AR, Dabbaghipour R, Sisakht M, Fallahi J. Partial reprogramming as a method for regenerating neural tissues in aged organisms. Cell Reprogram. 2024;26:10–23.

    Article  CAS  PubMed  Google Scholar 

  260. Neumann B, Segel M, Ghosh T, Zhao C, Tourlomousis P, Young A, Förster S, Sharma A, Chen CZ, Cubillos JF, Rawji KS, Chalut KJ, Franklin RJM. Myc determines the functional age state of oligodendrocyte progenitor cells. Nat Aging. 2021;1:826–37.

    Article  PubMed  Google Scholar 

  261. Rodríguez-Matellán A, Alcazar N, Hernández F, Serrano M, Ávila J. In vivo reprogramming ameliorates aging features in dentate gyrus cells and improves memory in mice. Stem Cell Rep. 2020;15:1056–66.

    Article  Google Scholar 

  262. Lu Y, Brommer B, Tian X, Krishnan A, Meer M, Wang C, Vera DL, Zeng Q, Yu D, Bonkowski MS, Yang JH, Zhou S, Hoffmann EM, Karg MM, Schultz MB, Kane AE, Davidsohn N, Korobkina E, Chwalek K, Rajman LA, Church GM, Hochedlinger K, Gladyshev VN, Horvath S, Levine ME, Gregory-Ksander MS, Ksander BR, He Z, Sinclair DA. Reprogramming to recover youthful epigenetic information and restore vision. Nature. 2020;588:124–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  263. Shen Y, Zaballa S, Bech X, Sancho-Balsells A, Díaz-Cifuentes C, Seyit-Bremer G, Ballasch I, Alcázar N, Alberch J, Abad M, Serrano M, Klein R. Giralt A and del Toro D. Expansion of the neocortex and protection from neurodegeneration by < em > in vivo transient reprogramming. bioRxiv. 2023:2023.11.27.568858.

  264. Seo JH, Lee MY, Yu JH, Kim MS, Song M, Seo CH. Kim HH and cho SR. In situ pluripotency factor expression promotes functional recovery from cerebral ischemia. Mol Ther. 2016;24:1538–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  265. Xu L, Ramirez-Matias J, Hauptschein M, Sun ED, Lunger JC, Buckley MT, Brunet A. Restoration of neuronal progenitors by partial reprogramming in the aged neurogenic niche. Nat Aging. 2024;4:546–67.

    Article  CAS  PubMed  Google Scholar 

  266. Miyamoto T, Furusawa C, Kaneko K. Pluripotency, differentiation, and reprogramming: A gene expression dynamics model with epigenetic feedback regulation. PLoS Comput Biol. 2015;11:e1004476.

    Article  PubMed  PubMed Central  Google Scholar 

  267. Mosteiro L, Pantoja C, Alcazar N, Marión RM, Chondronasiou D, Rovira M, Fernandez-Marcos PJ, Muñoz-Martin M, Blanco-Aparicio C, Pastor J, Gómez-López G, De Martino A, Blasco MA, Abad M, Serrano M. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science. 2016;354.

  268. Ohnishi K, Semi K, Yamamoto T, Shimizu M, Tanaka A, Mitsunaga K, Okita K, Osafune K, Arioka Y, Maeda T, Soejima H, Moriwaki H, Yamanaka S, Woltjen K, Yamada Y. Premature termination of reprogramming in vivo leads to cancer development through altered epigenetic regulation. Cell. 2014;156:663–77.

    Article  CAS  PubMed  Google Scholar 

  269. Abad M, Mosteiro L, Pantoja C, Cañamero M, Rayon T, Ors I, Graña O, Megías D, Domínguez O, Martínez D, Manzanares M, Ortega S, Serrano M. Reprogramming in vivo produces teratomas and iPS cells with totipotency features. Nature. 2013;502:340–5.

    Article  CAS  PubMed  Google Scholar 

  270. Ji J, Ng SH, Sharma V, Neculai D, Hussein S, Sam M, Trinh Q, Church GM, McPherson JD, Nagy A, Batada NN. Elevated coding mutation rate during the reprogramming of human somatic cells into induced pluripotent stem cells. Stem Cells. 2012;30:435–40.

    Article  CAS  PubMed  Google Scholar 

  271. Kosanke M, Osetek K, Haase A, Wiehlmann L, Davenport C, Schwarzer A, Adams F, Kleppa MJ, Schambach A, Merkert S, Wunderlich S, Menke S, Dorda M, Martin U. Reprogramming enriches for somatic cell clones with small-scale mutations in cancer-associated genes. Mol Ther. 2021;29:2535–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  272. Parras A, Vílchez-Acosta A, Desdín-Micó G, Picó S, Mrabti C, Montenegro-Borbolla E, Maroun CY, Haghani A, Brooke R, Del Carmen Maza M, Rechsteiner C, Battiston F, Branchina C, Perez K, Horvath S, Bertelli C. Sempoux C and Ocampo A. In vivo reprogramming leads to premature death linked to hepatic and intestinal failure. Nat Aging. 2023;3:1509–20.

    Article  PubMed  Google Scholar 

  273. Kim J, Kim S, Lee SY, Jo BK, Oh JY, Kwon EJ, Kim KT, Adpaikar AA, Kim EJ, Jung HS, Kim HR, Roe JS, Hong CP, Kim JK, Koo BK, Cha HJ. Partial in vivo reprogramming enables injury-free intestinal regeneration via autonomous Ptgs1 induction. Sci Adv. 2023;9:eadi8454.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Baldari S, Di Rocco G, Piccoli M, Pozzobon M, Muraca M, Toietta G. Challenges and strategies for improving the regenerative effects of mesenchymal stromal Cell-Based therapies. Int J Mol Sci. 2017;18.

  275. Ludke A, Li RK, Weisel RD. The rejuvenation of aged stem cells for cardiac repair. Can J Cardiol. 2014;30:1299–306.

    Article  PubMed  Google Scholar 

  276. Pettitt D, Holländer G, Brindley D. 718. Cell therapy commercialization: an assessment of translational barriers. Mol Ther. 2016;24.

  277. Zhang L, Yang F, Feng S. Allogeneic hematopoietic stem-cell transplantation for myelofibrosis. Ther Adv Hematol. 2020;11:2040620720906002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  278. Biddy BA, Kong W, Kamimoto K, Guo C, Waye SE, Sun T, Morris SA. Single-cell mapping of lineage and identity in direct reprogramming. Nature. 2018;564:219–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Yoo J, Chang Y, Kim H, Baek S, Choi H, Jeong GJ, Shin J, Kim H, Kim BS, Kim J. Efficient direct lineage reprogramming of fibroblasts into induced cardiomyocytes using nanotopographical cues. J Biomed Nanotechnol. 2017;13:269–79.

    Article  CAS  PubMed  Google Scholar 

  280. Yoo J, Noh M, Kim H, Jeon NL, Kim BS, Kim J. Nanogrooved substrate promotes direct lineage reprogramming of fibroblasts to functional induced dopaminergic neurons. Biomaterials. 2015;45:36–45.

    Article  CAS  PubMed  Google Scholar 

  281. Knappe N, Novak D, Weina K, Bernhardt M, Reith M, Larribere L, Hölzel M, Tüting T, Gebhardt C. Umansky V and Utikal J. Directed dedifferentiation using partial reprogramming induces invasive phenotype in melanoma cells. Stem Cells. 2016;34:832–46.

    Article  CAS  PubMed  Google Scholar 

  282. Huang Y, Wang T, López MEU, Hirano M, Hasan A, Shin SR. Recent advancements of human iPSC derived cardiomyocytes in drug screening and tissue regeneration. Microphysiol Syst. 2020;4:2.

    Article  PubMed  PubMed Central  Google Scholar 

  283. Lowenthal J, Sugarman J. Ethics and policy issues for stem cell research and pulmonary medicine. Chest. 2015;147:824–34.

    Article  PubMed  PubMed Central  Google Scholar 

  284. Volarevic V, Markovic BS, Gazdic M, Volarevic A, Jovicic N, Arsenijevic N, Armstrong L, Djonov V, Lako M, Stojkovic M. Ethical and safety issues of stem Cell-Based therapy. Int J Med Sci. 2018;15:36–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors declare that they have not use AI-generated work in this manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (32300675, 32400598), Sichuan Science and Technology Program (2022YFS0615, 2025ZNSFSC1019), Luzhou Science and Technology Program of China (2023SYF136, 2023JYJ019).

Author information

Authors and Affiliations

Authors

Contributions

Conception: Bingqing Xie and Huangfan Xie; manuscript draft: Sixiu Deng, Bingqing Xie, and Huangfan Xie; manuscript revision: Bingqing Xie, and Huangfan Xie. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Huangfan Xie or Bingqing Xie.

Ethics declarations

Conflicts of interest

The authors declare no conflicts of interest.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Deng, S., Xie, H. & Xie, B. Cell-based regenerative and rejuvenation strategies for treating neurodegenerative diseases. Stem Cell Res Ther 16, 167 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04285-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04285-7

Keywords