Skip to main content

Stem cell therapies for neurological disorders: current progress, challenges, and future perspectives

Abstract

Stem cell-based therapies have emerged as a promising approach for treating various neurological disorders by harnessing the regenerative potential of stem cells to restore damaged neural tissue and circuitry. This comprehensive review provides an in-depth analysis of the current state of stem cell applications in primary neurological conditions, including Parkinson’s disease (PD), Alzheimer’s disease (AD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), stroke, spinal cord injury (SCI), and other related disorders. The review begins with a detailed introduction to stem cell biology, discussing the types, sources, and mechanisms of action of stem cells in neurological therapies. It then critically examines the preclinical evidence from animal models and early human trials investigating the safety, feasibility, and efficacy of different stem cell types, such as embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), neural stem cells (NSCs), and induced pluripotent stem cells (iPSCs). While ESCs have been studied extensively in preclinical models, clinical trials have primarily focused on adult stem cells such as MSCs and NSCs, as well as iPSCs and their derivatives. We critically assess the current state of research for each cell type, highlighting their potential applications and limitations in different neurological conditions. The review synthesizes key findings from recent, high-quality studies for each neurological condition, discussing cell manufacturing, delivery methods, and therapeutic outcomes. While the potential of stem cells to replace lost neurons and directly reconstruct neural circuits is highlighted, the review emphasizes the critical role of paracrine and immunomodulatory mechanisms in mediating the therapeutic effects of stem cells in most neurological disorders. The article also explores the challenges and limitations associated with translating stem cell therapies into clinical practice, including issues related to cell sourcing, scalability, safety, and regulatory considerations. Furthermore, it discusses future directions and opportunities for advancing stem cell-based treatments, such as gene editing, biomaterials, personalized iPSC-derived therapies, and novel delivery strategies. The review concludes by emphasizing the transformative potential of stem cell therapies in revolutionizing the treatment of neurological disorders while acknowledging the need for rigorous clinical trials, standardized protocols, and multidisciplinary collaboration to realize their full therapeutic promise.

Introduction

Neurological disorders encompass a wide range of debilitating conditions that affect the central and peripheral nervous systems, leading to progressive damage and loss of neural tissue. These conditions include neurodegenerative illnesses, which are typified by the build-up of abnormal protein aggregates and the progressive loss of particular neuronal populations. Examples of these illnesses are Alzheimer’s disease (AD), Parkinson’s disease (PD), and Huntington’s disease (HD) [1, 2]. Other neurological conditions, such as multiple sclerosis (MS) and spinal cord injury (SCI), involve damage to the myelin sheath and axons, disrupting neural transmission and causing functional impairments [3, 4]. Cerebrovascular disorders, including stroke and traumatic brain injury (TBI), result in acute neural tissue damage and subsequent neuroinflammation, leading to long-term disability [5, 6]. Neurological disorders pose a significant burden on global health, affecting millions of individuals worldwide and leading to substantial healthcare costs and societal impact [7, 8].

Current therapeutic approaches for neurological disorders primarily focus on managing symptoms and slowing disease progression rather than addressing the underlying pathology. This is in part because the precise etiology of many neurological conditions remains unknown, limiting our ability to develop targeted disease-modifying therapies. Pharmacological treatments, such as dopaminergic medications for PD, cholinesterase inhibitors for AD, and immunomodulatory drugs for MS, provide symptomatic relief but often have limited efficacy and side effects [9,10,11]. Rehabilitation strategies aim to promote functional recovery and adaptations but do not directly restore lost neural tissue [12]. Surgical interventions, such as deep brain stimulation for PD, can alleviate specific symptoms but do not halt or reverse the neurodegenerative process [13]. While progress has been made in developing neuroprotective agents and gene therapies, their clinical translation has been challenging, and their long-term efficacy remains to be established [14, 15]. Given the limitations of current therapies, there is a pressing need for novel approaches that can effectively regenerate damaged neural tissue, replace lost neurons, and promote functional recovery in neurological disorders.

By utilizing stem cells' capacity for regeneration, stem cell-based therapies have become a viable option for treating the underlying pathophysiology of neurological illnesses. Because of their capacity to self-renew and differentiate into distinct cell types, stem cells are desirable in regenerative medicine [16]. Numerous stem cell types have been investigated for their potential as therapeutics for neurological illnesses, including induced pluripotent stem cells (iPSCs), neural stem cells (NSCs), mesenchymal stem cells (MSCs), and embryonic stem cells (ESCs) [17, 18]. These cells offer a variety of alternatives for cell-based therapeutics since they can be produced from various sources, including adult bone marrow, adipose tissue, embryonic tissue, and reprogrammed somatic cells [19, 20]. Stem cells have a variety of therapeutic applications in neurological illnesses, including immunomodulation, cell replacement, paracrine signaling, and stimulation of endogenous repair mechanisms [21,22,23]. Preclinical studies in animal models have demonstrated the ability of stem cells to differentiate into neuronal and glial lineages, integrate into host neural circuits, and promote functional recovery in various neurological conditions [24, 25]. However, the clinical translation of stem cell therapies faces numerous challenges, including optimizing cell manufacturing, delivery methods, and safety assessments [26].

This comprehensive review aims to provide an in-depth analysis of the current state of stem cell-based therapies for neurological disorders, focusing on the most recent advances and clinical applications. The review will discuss the biological properties and therapeutic mechanisms of different stem cell types, critically examine the preclinical and clinical evidence for their efficacy and safety, and highlight the challenges and future directions in the field. By synthesizing the latest research findings and expert opinions, this review seeks to inform researchers, clinicians, and stakeholders about the potential and limitations of stem cell therapies in revolutionizing the treatment of neurological disorders.

Stem cell basics

Stem cells are unspecialized cells with the unique ability to self-renew and differentiate into various cell types, making them a valuable tool for regenerative medicine [27]. Understanding stem cells' fundamental properties and mechanisms is crucial for their therapeutic application in neurological disorders. This section provides an overview of the different types of stem cells, their sources, and their mechanisms of action in neurological therapies.

Based on their potential for differentiation and developmental stage, stem cells can be categorized. All of the body's cell types can be produced by ESCs, pluripotent cells formed from the inner cell mass of blastocysts [28]. Nevertheless, there are hazards of tumor growth and ethical issues with using ESCs [29]. Adult stem cells are multipotent cells in bone marrow, adipose tissue, and the central nervous system. Examples of these tissues are MSCs and NSCs [30, 31]. Though less contentious and safer than ESCs, these cells have a more constrained capacity for differentiation [32]. Adult somatic cells are reprogrammed into a pluripotent state using particular transcription factors to create iPSCs [33]. iPSCs possess similar properties to ESCs but avoid the ethical issues associated with embryonic tissue use [34]. Other stem cell sources include perinatal tissues, such as umbilical cord blood and amniotic fluid, which contain a mix of multipotent stem cells [35].

Stem cells exert their therapeutic effects in neurological disorders through multiple mechanisms, broadly categorized into cell replacement, paracrine signaling, immunomodulation, and stimulation of endogenous repair processes.

  1. 1.

    Cell replacement: Stem cells can differentiate into specific neuronal and glial cell types, potentially replacing damaged or lost neural cells in neurological disorders [36]. For instance, dopaminergic neurons derived from stem cells can be transplanted into the striatum to replace degenerated neurons in Parkinson's disease [37]. However, the extent of cell replacement and functional integration of transplanted cells varies across different neurological conditions and requires further optimization [38].

  2. 2.

    Paracrine signaling: Stem cells secrete a wide range of bioactive molecules, including growth factors, cytokines, and extracellular vesicles, which can exert neuroprotective, anti-inflammatory, and regenerative effects on the host neural tissue [39, 40]. These paracrine factors can promote the survival and regeneration of endogenous neural cells, modulate the immune response, and enhance angiogenesis and neuroplasticity [41, 42]. The paracrine mechanisms of stem cells are believed to play a crucial role in their therapeutic efficacy, particularly in conditions where cell replacement alone may not be sufficient [43].

  3. 3.

    Immunomodulation: Neuroinflammation is a common feature of many neurological disorders, contributing to neural damage and hindering repair processes [44]. Stem cells, particularly MSCs, possess immunomodulatory properties that can regulate the immune response and create a more favorable environment for neural repair [45]. These cells can interact with various immune cells, such as T, B, and microglia, and modulate their activity through direct cell–cell contact and secretion of soluble factors [46, 47]. By attenuating neuroinflammation and promoting a pro-regenerative immune response, stem cells can indirectly support neural repair and functional recovery [48].

  4. 4.

    Stimulation of endogenous repair: To encourage the proliferation, differentiation, and integration of endogenous stem and progenitor cells into the injured neural tissue, stem cells can activate and mobilize these cells in the brain [49, 50]. Growth factors and chemokines that draw endogenous stem cells to the injury site and promote their survival and differentiation can be secreted to do this [51]. Furthermore, stem cells can expand the brain's neurogenic and angiogenic niches, improving the conditions for endogenous repair mechanisms [52].

The therapeutic mechanisms of stem cells in neurological disorders are complex and multifaceted, often involving a combination of cell replacement, paracrine signaling, immunomodulation, and stimulation of endogenous repair. The relative contribution of each mechanism may vary depending on the specific neurological condition, the type of stem cells used, and the route and timing of administration [53]. Understanding these mechanisms is crucial for optimizing stem cell-based therapies and developing targeted approaches for neurological disorders.

However cell-based therapy for neurological illnesses encounters various obstacles despite its considerable potential:

  • The potential of developing tumours, especially with pluripotent stem cells, is a concern [54]

  • Allogeneic cell transplants can be rejected by the immune system [55]

  • Cell survival and incorporation in the host tissue are restricted

  • The possibility of unregulated differentiation or movement [56]

  • There are ethical considerations related to the utilisation of embryonic stem cells [57]

  • Issues related to scalability and manufacture of cell products that meet clinical-grade standards [58].

There are questions about the safety of anything over a long period of time, and it is necessary to do additional studies to continue monitoring it [59].

These constraints highlight the significance of thorough preclinical testing and meticulous clinical trial design. Furthermore, it is imperative for regulatory frameworks to adapt in order to effectively tackle the distinctive obstacles presented by cell-based therapies [20].

Each type of stem cell has unique benefits and drawbacks when it comes to its use in neurological applications. ESCs possess a significant degree of adaptability, but they can give rise to ethical dilemmas and pose the possibility of developing tumours [60]. Adult stem cells, such as MSCs and NSCs, have a narrower range of cell types they can develop into, but they may present fewer safety risks [61]. iPSCs offer a means of obtaining cells that are particular to each patient, but they must undergo thorough analysis to guarantee their safety and effectiveness [31]. When selecting a cell type, it is important to thoroughly evaluate the individual neurological disease and therapeutic objectives [35].

Stem cell therapy in specific neurological diseases

Alzheimer’s disease

Amyloid-beta (Aβ) plaques and neurofibrillary tangles build up in Alzheimer's disease (AD), a progressive neurodegenerative condition that impairs memory, causes neuronal death and declines cognitive function [62]. Current pharmacological treatments, such as cholinesterase inhibitors and memantine, provide symptomatic relief but do not address the underlying pathology or halt disease progression [63]. Stem cell-based therapies have been proposed as a potential strategy to address multiple aspects of Alzheimer's disease pathology, including replacing lost neurons, providing neuroprotection, and modulating neuroinflammation [64].

Preclinical studies

Preclinical studies using animal models of AD have demonstrated the potential of various stem cell types, including MSCs, NSCs, and iPSCs, to ameliorate AD pathology and improve cognitive function. MSCs have been shown to reduce Aβ deposition, attenuate neuroinflammation, and promote neurogenesis and synaptic plasticity in AD mouse models [65,66,67]. NSCs derived from human fetal tissue or differentiated from pluripotent stem cells have been reported to differentiate into cholinergic neurons and integrate into the host brain, improving cognitive function in AD animal models [68, 69]. However, transplanted cells' long-term survival and functional integration remain challenging [70]. iPSC-derived neural cells have also shown promise in preclinical studies, with the advantage of allowing patient-specific and genetically corrected cell therapies [71, 72].

Clinical trials

To date, only a limited number of small-scale clinical trials have investigated the safety and feasibility of stem cell therapies in AD patients. A phase I trial using human umbilical cord blood-derived MSCs (hUCB-MSCs) demonstrated the safety and tolerability of repeated intravenous infusions in AD patients, with some evidence of stabilization of cognitive function [73]. Another phase I trial using autologous adipose-derived MSCs (ADSCs) showed safety and potential efficacy in slowing cognitive decline in mild to moderate AD patients [74]. However, these early-stage trials have limitations, such as small sample sizes, lack of placebo controls, and short follow-up periods, making it difficult to draw definitive conclusions about the efficacy of stem cell therapies in AD [75].

Challenges and future directions

Obstacles and prospects for the future although the preclinical results show promise, various hurdles must be overcome to successfully apply stem cell therapy in the clinical treatment of AD. These include enhancing the efficiency and quality of stem cells, enhancing the viability and effective integration of transplanted cells, and devising precise delivery techniques to specific brain regions impacted by AD [76]. Additionally, the optimal timing of intervention, the long-term safety and efficacy, and the potential need for repeated treatments need to be established through well-designed clinical trials [67].

Future directions in stem cell therapy for AD may involve using genetically modified stem cells to enhance their therapeutic properties, such as increased secretion of neurotrophic factors or Aβ-degrading enzymes [77]. Combining stem cell therapy with other therapeutic approaches, such as Aβ immunization or small molecule inhibitors of Aβ and tau pathology, may provide synergistic benefits [78]. 3D organoid models derived from patient-specific iPSCs may also facilitate drug screening and personalized treatment strategies [79].

Although stem cell therapy shows potential as a disease-modifying treatment for AD, additional research is required to tackle the obstacles and enhance the therapeutic strategy. Thorough preclinical investigations and well-planned clinical trials are necessary to determine stem cell treatments' safety, effectiveness, and long-term advantages in Alzheimer's disease. However, the complex and multifaceted nature of AD presents significant challenges for developing effective cell replacement therapies. AD involves widespread neuronal loss, synaptic dysfunction, protein aggregation, and vascular abnormalities across multiple brain regions. Simple cell replacement is unlikely to address all of these pathological features. Additional research is required to determine if stem cell approaches can tackle the numerous obstacles presented by AD's complexity and enhance therapeutic strategies [80].

Parkinson’s disease

Parkinson's disease (PD) is a degenerative neurological condition that gradually causes the death of dopaminergic neurons in the substantia nigra pars compacta (SNpc). This leads to motor symptoms, including tremors, stiffness, and slowness of movement [81]. Current treatments, including dopaminergic medications and deep brain stimulation, provide symptomatic relief but do not address the underlying neuronal loss or halt disease progression [82]. Stem cell-based therapies aim to replace lost dopaminergic neurons and restore motor function in PD [83]. It is important to note that Parkinson's disease is not simply characterized by the loss of dopaminergic neurons. The pathophysiology is complex, involving multiple neurotransmitter systems, protein aggregation, neuroinflammation, and dysfunction of various neural circuits. For stem cell therapies to be truly disease-modifying or curative, they would need to address these multiple aspects of PD pathology. Recent research has highlighted additional challenges, including the potential spread of alpha-synuclein pathology to transplanted cells and the need to restore broader neural circuit function beyond dopamine replacement [84].

Preclinical studies

Preclinical studies using animal models of PD have demonstrated the potential of various stem cell types, particularly ESCs and iPSCs, to differentiate into dopaminergic neurons and improve motor function. Transplantation of human ESC-derived dopaminergic neurons into the striatum of PD animal models has shown survival, integration, and functional recovery [85, 86]. Similarly, iPSC-derived dopaminergic neurons have demonstrated the ability to engraft, innervate the host striatum, and ameliorate motor deficits in PD models [87, 88]. However, challenges such as variability in differentiation efficiency, graft survival, and potential tumorigenicity need to be addressed [89]. PD models have also investigated MSCs for their neuroprotective and immunomodulatory properties [90, 91].

Clinical trials

Several clinical trials have investigated the safety and efficacy of stem cell therapies in PD patients. Early trials using fetal ventral mesencephalic (FVM) tissue grafts demonstrated variable outcomes, with some patients showing long-term clinical benefits and others developing graft-induced dyskinesias [92, 93]. More recently, clinical trials using human ESC-derived dopaminergic progenitors have shown promise. A phase 1/2 trial reported the safety and survival of transplanted cells in PD patients, with some evidence of motor improvement [94]. An ongoing phase 1 trial (NCT03119636) investigates the safety and efficacy of human ESC-derived dopaminergic progenitors in PD patients [95]. Clinical trials using autologous iPSC-derived dopaminergic neurons are also in the planning stages [96].

Challenges and future directions

While stem cell therapy for PD has made significant progress, several challenges remain. These include optimizing the differentiation and purification of dopaminergic neurons, ensuring graft survival and functional integration, and minimizing the risk of graft-induced dyskinesias [97]. Strategies to enhance graft survival, such as co-transplantation with supportive cell types or neuroprotective agents, are being explored [95]. The development of standardized protocols for cell manufacturing and quality control is also essential for the reproducibility and scalability of stem cell therapies [98]. Future directions in stem cell therapy for PD may involve gene editing technologies to correct disease-causing mutations in patient-specific iPSCs [99]. Cell encapsulation or bioengineered scaffolds may improve graft survival and integration [100]. Combinatorial approaches, such as the co-administration of neurotrophic factors or the use of neuroprotective agents, may enhance the therapeutic efficacy of stem cell therapies [101].

Stem cell therapy shows potential as a disease-modifying treatment for PD, aiming to replace lost dopaminergic neurons and restore motor function. Although there have been promising findings in preclinical studies and early clinical trials, additional research is required to tackle the obstacles and enhance the therapeutic approach. Continuing and upcoming clinical trials will offer valuable knowledge regarding the safety, effectiveness, and long-term advantages of stem cell therapies in PD.

Multiple sclerosis

Multiple sclerosis (MS) is a long-lasting inflammatory condition of the CNS that involves the immune system attacking and damaging the protective covering of nerve fibers called myelin. This damage results in neurological problems and impairment [102]. Current therapies for MS primarily focus on immunomodulation and symptom management but do not effectively promote remyelination or prevent progressive neurodegeneration [103]. Stem cell-based therapies aim to promote remyelination, provide neuroprotection, and modulate the immune response in MS. Among neurological conditions, multiple sclerosis (MS) stands out as having the most advanced clinical applications of stem cell therapy. Autologous hematopoietic stem cell transplantation (aHSCT) is now routinely used in medical centers worldwide to treat aggressive forms of MS. This approach aims to 'reset' the immune system and halt disease progression [104].

Preclinical studies

Preclinical studies using animal models of MS, such as experimental autoimmune encephalomyelitis (EAE), have demonstrated the potential of various stem cell types to promote remyelination and ameliorate disease progression. Transplantation of NSCs or oligodendrocyte progenitor cells (OPCs) derived from ESCs or iPSCs has shown the ability to differentiate into mature oligodendrocytes, promote remyelination, and improve functional outcomes in EAE models [105, 106]. MSCs have also been extensively studied in MS models for their immunomodulatory and neuroprotective properties [107]. MSCs have been shown to reduce neuroinflammation, suppress autoreactive T cells, and promote the generation of regulatory T cells, leading to improved clinical outcomes in EAE [108, 109].

Clinical trials

Several clinical trials have investigated the safety and efficacy of stem cell therapies in MS patients. aHSCT has been explored as a potential treatment for aggressive forms of MS to reset the immune system and halt disease progression. Comparing aHSCT to disease-modifying therapies found that aHSCT was superior in preventing disease progression and achieving sustained improvement in neurological function. Long-term follow-up studies have shown that a significant proportion of patients remain free from disease activity for 5 years or more after treatment [110]. While some studies have shown promising results, with long-term stabilization or improvement of disability in a subset of patients, the procedure is associated with significant risks. It is currently reserved for select patients with highly active disease [111, 112]. Clinical trials using MSCs have also been conducted in MS patients, primarily focusing on safety and feasibility [113]. Intravenous administration of autologous MSCs is well-tolerated, with evidence of potential efficacy in reducing inflammatory activity and promoting neuroprotection [102, 114]. However, more extensive randomized controlled trials are needed to establish the long-term safety and efficacy of MSC-based therapies in MS.

Challenges and future directions

While stem cell therapies hold promise for the treatment of MS, several challenges need to be addressed. One of the main challenges is ensuring the survival, differentiation, and functional integration of transplanted cells in the host CNS [115]. Strategies to enhance graft survival and promote targeted migration to sites of demyelination are being explored [110]. Another challenge is the potential for graft rejection or the development of secondary autoimmunity [116]. The use of autologous or genetically modified stem cells and the development of improved immunosuppressive regimens may help mitigate these risks [117]. Future directions in stem cell therapy for MS may involve using gene editing technologies to create "off-the-shelf" cell products with enhanced remyelination capacity or immunomodulatory properties [118]. Developing biomaterials and tissue engineering approaches to create scaffolds that support cell survival and guide axonal regeneration is also an active area of research [119]. Combination therapies that target multiple aspects of MS pathology, such as neuroinflammation, oxidative stress, and mitochondrial dysfunction, may enhance the therapeutic potential of stem cell transplantation [120].

Stem cell-based therapies for MS have shown promising results in preclinical studies and early clinical trials. While challenges remain, advances in cell manufacturing, genetic engineering, and biomaterial science are expected to improve the safety, efficacy, and accessibility of stem cell therapies for MS in the future. Further research and well-designed clinical trials are needed to establish the optimal therapeutic approach and long-term benefits of stem cell transplantation in MS. The success of aHSCT in MS has led to its inclusion in treatment guidelines for highly active relapsing–remitting MS that is refractory to conventional therapies. However, patient selection is crucial, as the procedure carries risks and is most beneficial for younger patients with active inflammatory disease. Ongoing research is focused on optimizing aHSCT protocols, reducing treatment-related risks, and exploring its potential in progressive forms of MS. Additionally, other stem cell approaches, such as mesenchymal stem cell therapies, are being investigated for their potential neuroprotective and regenerative properties in MS [121].

Stroke

Stroke is a leading cause of death and disability worldwide, characterized by the sudden loss of blood supply to the brain, resulting in neuronal damage and functional impairments [122]. Current treatments for stroke primarily focus on restoring blood flow and providing supportive care but do not effectively address the long-term neurological deficits [123]. Stem cell-based therapies promote neuronal repair, modulate inflammation, and enhance functional recovery in stroke [124].

Preclinical studies

Animal research investigating stroke has shown that different types of stem cells, such as MSCs, NSCs, and iPSCs, can enhance neuronal repair and functional outcomes. Studies have demonstrated that the transplantation of MSCs can decrease the extent of tissue damage caused by a lack of blood supply, regulate the inflammation of nerves, and improve the growth of new nerve cells and blood vessels in animal stroke models [125, 126]. NSCs, either from fetal tissue or generated from induced iPSCs, have shown the capacity to move towards the location of injury, transform into nerve cells and support cells, and enhance the restoration of function in stroke models [127, 128]. Nevertheless, the precise timing, method, and amount of stem cell administration and the sustained viability and incorporation of transplanted cells continue to be significant obstacles [129].

Clinical trials

Several clinical trials have investigated the safety and feasibility of stem cell therapies in stroke patients. A meta-analysis of early-phase clinical trials using MSCs in ischemic stroke patients reported a favorable safety profile and potential improvements in functional outcomes [130]. However, the efficacy of MSC transplantation in stroke remains to be established in larger, randomized controlled trials. The MASTERS trial, a phase 2 study of intravenous administration of bone marrow-derived MSCs in acute ischemic stroke patients, showed no significant improvement in functional outcomes at 90 days compared to placebo [128]. More recently, the TREASURE trial, a phase 2/3 study of intravenous administration of umbilical cord blood-derived MSCs in acute ischemic stroke patients, also failed to demonstrate a significant improvement in functional outcomes at 90 days [131]. These results highlight the need for further optimization of stem cell therapies for stroke, including the selection of patients most likely to benefit, the timing and route of administration, and the potential for combination therapies [127].

Challenges and future directions

While stem cell therapies hold promise for the treatment of stroke, several challenges need to be addressed. One of the main challenges is the limited survival and engraftment of transplanted cells in the ischemic brain [132]. Strategies to enhance cell survival, such as preconditioning or genetic modification of stem cells, are being explored [133]. Another challenge is the potential for off-target effects or the development of adverse events, such as tumorigenesis or stroke-associated infection [134]. The use of highly purified and well-characterized cell populations and rigorous safety monitoring will be essential for the clinical translation of stem cell therapies for stroke [135]. Future directions in stem cell therapy for stroke may involve using biomaterials and tissue engineering approaches to create a supportive microenvironment for transplanted cells and enhance their survival and differentiation [136]. Developing cell-free approaches, such as using extracellular vesicles or exosomes derived from stem cells, may also provide a more scalable and safe alternative to cell transplantation [137]. Combinatorial approaches, such as the co-administration of neuroprotective agents or the use of rehabilitation therapies, may enhance the therapeutic efficacy of stem cell transplantation [138].

While stem cell-based therapies for stroke have shown promising results in preclinical studies, the clinical translation of these approaches has been challenging. Further research is needed to optimize the therapeutic approach, including selecting the most appropriate stem cell type, the timing and route of delivery, and the potential for combination therapies. Well-designed clinical trials with larger sample sizes and more extended follow-up periods will be essential to establish the safety and efficacy of stem cell therapies for stroke.

Amyotrophic lateral sclerosis

Muscle weakening, paralysis, and eventually death are the results of selective motor neuron loss in the brain and spinal cord that characterizes amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative illness [139]. There are currently just a few ALS treatments available, and they mainly concentrate on supportive care and symptom control [140]. The goals of stem cell-based treatments for ALS are to reduce neuroinflammation, restore damaged motor neurons, and offer neuroprotection [141].

Preclinical studies

Preclinical studies using animal models of ALS, such as the SOD1 transgenic mouse model, have demonstrated the potential of various stem cell types to delay disease progression and extend survival. Transplantation of NSCs or motor neuron progenitors derived from ESCs or iPSCs has been shown to integrate into the spinal cord, form synaptic connections with host neurons, and improve motor function in ALS models [141, 142]. MSCs have also been extensively studied in ALS models for their immunomodulatory and neuroprotective properties [143]. Intrathecal or intravenous administration of MSCs has been shown to reduce neuroinflammation, protect against motor neuron loss, and prolong survival in ALS mice [144, 145]. However, the long-term survival and efficacy of transplanted cells in the diseased microenvironment of ALS remain significant challenges [146].

Clinical trials

Several early-phase clinical trials have investigated the safety and feasibility of stem cell therapies in ALS patients. Intraspinal transplantation of fetal spinal cord-derived NSCs in ALS patients is safe and well-tolerated, with some evidence of potential efficacy in slowing disease progression [147, 148]. However, a follow-up phase 2 trial did not significantly improve functional outcomes or survival compared to placebo [149]. Intrathecal administration of autologous MSCs has also been explored in ALS patients, focusing on safety and tolerability [137, 150]. While these early trials have provided proof-of-concept for the feasibility of stem cell transplantation in ALS, more extensive randomized controlled trials are needed to establish the efficacy of these approaches.

Challenges and future directions

Despite the promising preclinical results, the clinical translation of stem cell therapies for ALS faces several challenges. One of the main challenges is the complex and multifactorial nature of ALS pathogenesis, which may limit the therapeutic efficacy of cell replacement alone [151]. Strategies to enhance the survival, integration, and function of transplanted cells in the hostile microenvironment of ALS are being explored, such as the co-transplantation of supportive glial cells or the use of neuroprotective factors [152]. Another challenge is the potential for immune rejection or the development of adverse events, such as graft-induced dyskinesias or tumorigenesis [153]. The use of autologous or genetically modified stem cells and improved immunosuppressive regimens may help mitigate these risks [154].

Future directions in stem cell therapy for ALS may involve gene editing technologies to correct ALS-causing mutations in patient-specific iPSCs, which could then be differentiated into healthy motor neurons for transplantation [155]. Using biomaterials and tissue engineering approaches to create scaffolds that support cell survival and guide axonal regeneration is also an active area of research [156]. Combinatorial approaches, such as the co-administration of neuroprotective agents or the use of anti-inflammatory drugs, may enhance the therapeutic potential of stem cell transplantation [157]. Developing novel delivery methods, such as intramuscular or intravascular administration of stem cells, may provide a less invasive and more scalable approach for cell therapy in ALS [158].

Although preclinical research on stem cell-based therapy for ALS has yielded encouraging findings, the practical application of these strategies has been complex. More studies are required on the most suitable stem cell type, administration timing and route, and the possibility of combination therapies to optimize the therapeutic strategy. More significant sample numbers and extended follow-up periods in carefully planned clinical trials will be necessary to confirm the safety and effectiveness of stem cell treatments for ALS. Additionally, the development of successful stem cell-based treatments for this debilitating illness will depend on a deeper comprehension of the underlying mechanisms of ALS pathogenesis and the interactions between transplanted cells and the host milieu.

Huntington’s disease

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by a trinucleotide repeat expansion in the huntingtin gene, leading to the production of a mutant huntingtin protein that causes progressive neuronal loss and dysfunction, particularly in the striatum and cortex [159]. Current treatments for HD are limited and primarily focus on managing symptoms, such as chorea and psychiatric disturbances [160]. Stem cell-based therapies aim to replace lost neurons, provide neuroprotection, and modulate neuroinflammation in HD [161].

Preclinical studies

Preclinical studies using animal models of HD, such as the R6/2 and YAC128 transgenic mouse models, have demonstrated the potential of various stem cell types to improve motor function, reduce neuronal loss, and extend survival. Transplantation of fetal striatal tissue or NSCs derived from ESCs or iPSCs has been shown to integrate into the striatum, form synaptic connections with host neurons, and ameliorate motor deficits in HD mice [162, 163]. MSCs have also been explored in HD models for their immunomodulatory and neuroprotective properties [164]. Intrastriatal or intravenous administration of MSCs has been shown to reduce neuroinflammation, increase neurotrophic factor levels, and improve motor function in HD mice [165, 166]. However, the long-term survival and efficacy of transplanted cells in the diseased microenvironment of HD remain significant challenges [167].

Clinical trials

To date, few clinical trials have investigated the safety and feasibility of stem cell therapies in HD patients. A phase 1 trial of fetal striatal tissue transplantation in HD patients demonstrated the safety and feasibility of the approach, with some evidence of graft survival and clinical benefit [168]. However, a follow-up study found that the transplanted cells developed HD-like pathology over time, suggesting that cell replacement alone may not halt disease progression [169]. Recently, a phase 1/2 trial of intrastriatal transplantation of human ESC-derived neural progenitors in HD patients has been initiated (NCT03252080) [170]. This trial aims to assess the approach's safety, tolerability, and preliminary efficacy, with results expected in the coming years.

Challenges and future directions

While stem cell-based therapies for HD hold promise, several challenges must be addressed for successful clinical translation. One of the main challenges is the potential for transplanted cells to acquire HD-related pathology over time due to the presence of the mutant huntingtin protein in the host environment [171]. Strategies to mitigate this risk, such as genetically corrected autologous iPSCs or the co-transplantation of neuroprotective factors, are being explored [172]. Another challenge is the need for targeted cell delivery to the affected brain regions, as widespread neuronal loss and circuit dysfunction occur in HD [173]. Developing advanced imaging techniques and stereotactic surgery methods may help guide precise cell transplantation [154]. Future directions in stem cell therapy for HD may involve gene editing technologies, such as CRISPR–Cas9, to correct the HTT mutation in patient-specific iPSCs, which could then be differentiated into healthy striatal neurons for transplantation [155]. Using biomaterials and tissue engineering approaches to create scaffolds that support cell survival and guide axonal regeneration is also an active area of research [156]. Combinatorial approaches, such as the co-administration of neuroprotective agents or the use of anti-inflammatory drugs, may enhance the therapeutic potential of stem cell transplantation [174]. Developing novel delivery methods, such as intracerebroventricular or intrathecal administration of stem cells, may provide a less invasive and more widespread approach for cell therapy in HD [175].

In summary, whereas preclinical research on stem cell-based treatments for Huntington's disease has yielded encouraging outcomes, the clinical application of these strategies is still in its infancy. More studies are required on the most suitable stem cell type, administration timing and route, and the possibility of combination therapies to optimize the therapeutic strategy. To prove that stem cell therapies for HD are safe and effective, well-designed clinical trials with bigger sample sizes and longer follow-up times will be necessary. Additionally, the development of successful stem cell-based treatments for this debilitating illness will depend on a deeper comprehension of the molecular mechanisms driving HD pathogenesis and the interactions between transplanted cells and the host milieu.

Spinal cord injury

A severe disorder known as spinal cord injury (SCI) causes the loss of motor and sensory function below the site of the damage, which frequently leads to permanent paralysis and impairment [176]. The main goals of current SCI treatments are to stabilize the spine, stop more injury, and encourage recovery [177]. The goals of stem cell-based treatments for spinal cord injury (SCI) include glia and missing neuron replacement, axonal regeneration, and inflammation response modulation [178].

Preclinical studies

The potential of different types of stem cells to support functional recovery and regeneration has been proven in preclinical research utilizing animal models of spinal cord injury. It has been demonstrated that transplanting NSCs or neural progenitor cells (NPCs) derived from ESCs or iPSCs into the injured spinal cord can improve motor function in rodent and primate models of SCI by promoting the differentiation of neurons and glia and forming synaptic connections with host neurons. Since MSCs exhibit immunomodulatory, neuroprotective, and pro-angiogenic qualities, they have also been the subject of substantial research in SCI models [179]. It has been demonstrated that administering MSCs intravenously or intraspinally to SCI mice can decrease inflammation, encourage tissue sparing, and improve functional recovery [180, 181]. Nonetheless, there are still significant obstacles to overcome, including the best time, method, and dosage for delivering stem cells and the integration and long-term survival of transplanted cells [182].

Clinical trials

Several clinical trials have investigated the safety and feasibility of stem cell therapies in SCI patients. A systematic review and meta-analysis of clinical trials using MSCs in SCI patients found no serious adverse events related to cell transplantation and some evidence of functional improvement [183]. However, the included studies were small, heterogeneous, and lacked appropriate controls, highlighting the need for more extensive, well-designed trials to establish the efficacy of MSC therapy in SCI [184]. A phase 2 trial of intramedullary transplantation of human ESC-derived oligodendrocyte progenitor cells in subacute SCI patients (NCT02302157) has recently been completed, with results pending publication [185]. Other ongoing or planned trials investigate the safety and efficacy of various stem cell types in SCI patients, including NSCs, NPCs, and autologous bone marrow-derived MSCs [186].

Challenges and future directions

While stem cell-based therapies for SCI hold promise, several challenges must be addressed for successful clinical translation. One of the main challenges is the complex and dynamic nature of the injury microenvironment, which may limit the survival, differentiation, and integration of transplanted cells [187]. Strategies to enhance cell survival and promote targeted differentiation, such as co-delivering neuroprotective factors or biomaterials and tissue engineering approaches, are being explored [188, 189]. Another challenge is the potential for adverse events, such as neuropathic pain, autonomic dysreflexia, or tumor formation, following stem cell transplantation [190]. Careful patient selection, rigorous safety monitoring, and long-term follow-up will be essential to mitigate these risks [184].

Future directions in stem cell therapy for SCI may involve gene editing technologies to engineer stem cells with enhanced regenerative properties, such as increased neurotrophic factor secretion or improved myelination capacity [191]. The development of advanced biomaterials and tissue engineering approaches to create scaffolds that mimic the natural extracellular matrix and guide axonal regeneration is also an active area of research [154]. Combinatorial approaches, such as the co-administration of rehabilitation therapy or the use of electrical stimulation, may enhance the therapeutic potential of stem cell transplantation [192]. Additionally, identifying reliable biomarkers and imaging techniques to monitor the survival, differentiation, and integration of transplanted cells in vivo will be critical for optimizing and individualizing stem cell therapies for SCI [193].

Stem cell-based therapies for SCI have shown promising results in preclinical studies, with growing evidence of safety and feasibility in early clinical trials. However, further research is needed to optimize the therapeutic approach, including selecting the most appropriate stem cell type, the timing and route of delivery, and the potential for combination therapies. Well-designed, randomized controlled trials with larger sample sizes, longer follow-up periods, and standardized outcome measures will be essential to establish the efficacy of stem cell therapies for SCI. Additionally, a deeper understanding of the molecular mechanisms underlying SCI pathophysiology and the interactions between transplanted cells and the host microenvironment will be critical for developing safe and effective stem cell-based therapies for this devastating condition.

Traumatic brain injury

Traumatic brain injury (TBI) is a leading cause of death and disability worldwide, resulting from sudden physical damage to the brain due to external forces, such as falls, vehicle accidents, or violence [194]. The primary injury initiates a cascade of secondary injury mechanisms, including neuroinflammation, oxidative stress, excitotoxicity, and apoptosis, leading to progressive neuronal loss and dysfunction [195]. Current treatments for TBI primarily focus on minimizing secondary injury, managing intracranial pressure, and providing rehabilitation to promote functional recovery [196]. However, there are no effective therapies to reverse the damage and restore function in chronic TBI patients [197]. Stem cell-based therapies aim to replace lost neurons and glia, modulate the inflammatory response, and promote neurogenesis and angiogenesis in TBI [198].

Preclinical studies

Preclinical studies using animal models of TBI have demonstrated the potential of various stem cell types, including neural stem/progenitor cells (NSPCs), MSCs, and HSCs, to promote functional recovery after TBI. Transplantation of NSPCs derived from ESCs or iPSCs into the injured brain has been shown to differentiate into neurons and glia, form synaptic connections with host neurons, and improve cognitive and motor function in rodent models of TBI [199, 200]. MSCs have also been extensively studied in TBI models for their immunomodulatory, neuroprotective, and pro-angiogenic properties [201]. Intravenous or intracerebral administration of MSCs has been shown to reduce inflammation, promote neurogenesis and angiogenesis, and enhance functional recovery in TBI animals [202, 203]. HSCs mobilized from the bone marrow have been shown to migrate to the injured brain, differentiate into microglia and neurons, and improve cognitive function in rodent models of TBI [204]. While preclinical studies of stem cell therapies for TBI have shown promise, significant challenges remain in translating these approaches to clinical practice. Further research is needed to optimize cell types, delivery methods, and timing of intervention. Importantly, the complex and heterogeneous nature of TBI may require combinatorial approaches rather than relying solely on cell replacement strategies.

Clinical trials

Several early-phase clinical trials have investigated the safety and feasibility of stem cell therapies in TBI patients. A phase 1/2a study of intravenous administration of autologous bone marrow-derived mononuclear cells (BMMNCs) in acute severe TBI patients demonstrated safety and a trend towards improved neurological outcomes [205]. Another phase 1 study of intracerebral transplantation of human NSCs in chronic TBI patients showed safety and feasibility, with some evidence of improved neurological function [206]. However, more extensive randomized controlled trials are needed to establish the efficacy of these approaches in improving functional outcomes and quality of life in TBI patients [207].

Challenges and future directions

While stem cell-based therapies for TBI hold promise, several challenges must be addressed for successful clinical translation. One of the main challenges is the heterogeneity of TBI, which can vary in terms of the mechanism, location, and severity of injury, as well as the age and comorbidities of the patient [208]. Developing personalized stem cell therapies tailored to each patient's specific needs may be necessary to maximize therapeutic efficacy [209]. Another challenge is the potential for adverse events, such as seizures, infection, or tumorigenesis, following stem cell transplantation [190]. Careful patient selection, rigorous safety monitoring, and long-term follow-up will be essential to mitigate these risks [210].

Future directions in stem cell therapy for TBI may involve gene editing technologies to enhance the regenerative properties of transplanted cells, such as overexpressing neurotrophic factors or anti-inflammatory cytokines [211]. Research is also being done using biomaterials and tissue engineering techniques to make scaffolds that resemble the extracellular matrix seen in nature and offer a favorable environment for cell survival and development [212]. The therapeutic potential of stem cell transplantation may be increased by combinatorial techniques, including co-administration of neuroprotective drugs, neurorestorative treatments, or rehabilitation [213]. Furthermore, developing noninvasive imaging modalities like PET and MRI to track the migration, survival, and differentiation of transplanted cells in vivo would be essential for customizing and streamlining stem cell treatments for traumatic brain injury [214].

Stem cell-based therapies for TBI have shown promising results in preclinical studies, with early evidence of safety and feasibility in clinical trials. However, further research is needed to optimize the therapeutic approach, including selecting the most appropriate stem cell type, the timing and route of delivery, and the potential for combination therapies. Well-designed, randomized controlled trials with larger sample sizes, longer follow-up periods, and standardized outcome measures will be essential to establish the efficacy of stem cell therapies for TBI. Additionally, a deeper understanding of the complex pathophysiology of TBI and the mechanisms underlying the therapeutic effects of stem cells will be critical for developing safe and effective regenerative medicine approaches for this devastating condition.

Epilepsy

Repeated, unprovoked seizures are a hallmark of epilepsy, a chronic neurological illness caused by abnormally high levels of aberrant brain neuronal activity [215]. Even though antiepileptic medications (AEDs) are the cornerstone of epilepsy treatment, over one-third of patients still do not respond to medication [216]. Intending to reestablish the proper balance between excitement and inhibition in the epileptic brain, stem cell-based therapies have become a viable adjunctive or alternative therapeutic option for drug-resistant epilepsy [217].

Preclinical studies

Preclinical studies using animal models of epilepsy have demonstrated the potential of various stem cell types, particularly GABAergic interneuron progenitors and MSCs, to suppress seizures and modify the underlying disease pathology. Transplantation of GABAergic interneuron progenitors derived from ESCs or iPSCs into the hippocampus or other seizure-prone regions has been shown to engraft, differentiate into functional GABAergic interneurons, and reduce seizure frequency and severity in rodent models of epilepsy [218, 219]. These effects are mediated by the synaptic integration of the transplanted cells into the host circuitry and the enhancement of inhibitory neurotransmission [220]. MSCs have also shown promise in preclinical epilepsy models, exerting anticonvulsant and neuroprotective effects through the secretion of neurotrophic factors and the modulation of inflammatory responses [221, 222]. However, the long-term efficacy and safety of stem cell therapies in epilepsy remain to be established [223].

Clinical trials

To date, few clinical trials have investigated the safety and efficacy of stem cell therapies in epilepsy patients. A phase 1 trial of autologous bone marrow-derived mononuclear cells (BMMNCs) administered intravenously in children with refractory epilepsy demonstrated safety and feasibility, with some evidence of reduced seizure frequency [224]. Another pilot study of intracerebral transplantation of autologous BMMNCs in adult patients with drug-resistant mesial temporal lobe epilepsy also showed safety and potential efficacy in reducing seizure frequency [225]. However, these early-stage trials are limited by small sample sizes, lack of control groups, and short follow-up periods, highlighting the need for larger, well-designed, randomized controlled trials to establish the efficacy of stem cell therapies in epilepsy [226].

Challenges and future directions

While stem cell-based therapies for epilepsy hold promise, several challenges must be addressed for successful clinical translation. One of the main challenges is the complex and multifactorial nature of epilepsy, which may require tailored stem cell therapies targeting specific epileptogenic mechanisms in each patient [227]. Another challenge is the potential for adverse events, such as graft rejection, tumor formation, or worsening of seizures, following stem cell transplantation [228]. Careful patient selection, rigorous safety monitoring, and long-term follow-up will be essential to mitigate these risks [210].

Future directions in stem cell therapy for epilepsy may involve gene editing technologies to create stem cell-derived GABAergic interneurons with enhanced anticonvulsant properties or reduced immunogenicity [229]. The development of advanced delivery methods, such as stereotactic surgery or convection-enhanced delivery, to achieve targeted and controlled transplantation of stem cells into the epileptic focus is also an active area of research [230]. Combinatorial approaches, such as the co-administration of neuroprotective agents or the use of gene therapy to overexpress seizure-suppressing molecules, may enhance the therapeutic potential of stem cell transplantation [231]. Additionally, identifying reliable biomarkers and advanced neuroimaging techniques to guide patient selection, monitor the fate of transplanted cells, and assess the efficacy of stem cell therapies in vivo will be critical for optimizing and individualizing stem cell-based treatments for epilepsy [232].

Stem cell-based therapies for epilepsy have shown promise in preclinical studies, with some evidence of safety and feasibility in early clinical trials. However, further research is needed to optimize the therapeutic approach, including selecting the most appropriate stem cell type, the timing and route of delivery, and the potential for combination therapies. Well-designed, randomized controlled trials with larger sample sizes, longer follow-up periods, and standardized outcome measures will be essential to establish the efficacy of stem cell therapies for epilepsy. Additionally, a deeper understanding of the complex pathophysiology of epilepsy and the mechanisms underlying the therapeutic effects of stem cells will be critical for developing safe and effective regenerative medicine approaches for this challenging neurological disorder.

Other emerging applications

In addition to the neurological disorders discussed above, stem cell-based therapies have shown potential for the treatment of various other neurological conditions, such as cerebral palsy, autism spectrum disorder (ASD), and peripheral nerve injuries.

Cerebral palsy

A set of lifelong mobility abnormalities known as cerebral palsy are brought on by harm to the developing brain and first manifest in early childhood [233]. Current treatments for cerebral palsy primarily focus on managing symptoms and improving function through physical therapy, occupational therapy, and medications [234]. Stem cell-based therapies, particularly umbilical cord blood (UCB) cells and MSCs, have shown promise in preclinical and early clinical studies for cerebral palsy [235]. These cells have been shown to exert neuroprotective, anti-inflammatory, and pro-angiogenic effects, promoting brain repair and functional recovery [236, 237]. However, more extensive randomized controlled trials are needed to establish stem cell therapies' efficacy and long-term safety for cerebral palsy [238].

Autism spectrum disorder

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and interaction, along with restricted and repetitive patterns of behavior [239]. While behavioral and educational interventions are the mainstay of treatment for ASD, there are no effective pharmacological therapies to address the core symptoms [240]. Stem cell-based therapies, particularly MSCs and NSCs, have shown potential in preclinical studies to modulate the immune system, promote synaptic plasticity, and improve behavioral outcomes in animal models of ASD [241, 242]. A few small clinical studies have investigated the safety and feasibility of stem cell therapies in ASD patients, with some evidence of improved behavioral and cognitive function [243, 244]. However, these studies are limited by small sample sizes, lack of control groups, and short follow-up periods, highlighting the need for larger, well-designed clinical trials to establish the efficacy and safety of stem cell therapies for ASD [245].

Peripheral nerve injuries

Peripheral nerve injuries caused by trauma, surgery, or disease can lead to sensory and motor deficits, neuropathic pain, and reduced quality of life [246]. Current treatments for peripheral nerve injuries primarily focus on surgical repair, physical therapy, and pain management [246]. Stem cell-based therapies, particularly Schwann cells, MSCs, and adipose-derived stem cells (ADSCs), have shown promise in preclinical studies to promote nerve regeneration, remyelination, and functional recovery [247,248,249]. These cells can be transplanted directly into the injured nerve or delivered through nerve guidance conduits or other biomaterial scaffolds [250]. A few early-stage clinical trials have investigated the safety and feasibility of stem cell therapies for peripheral nerve injuries, with some evidence of improved sensory and motor function [176, 251]. However, further research is needed to optimize the therapeutic approach and establish stem cell therapies' long-term efficacy and safety for peripheral nerve injuries [247].

Challenges and future directions

While stem cell-based therapies for these neurological disorders hold promise, several challenges must be addressed for successful clinical translation. These include the heterogeneity of the patient population, the complex and multifactorial nature of the underlying pathology, and the potential for adverse events following stem cell transplantation [252]. Future directions may involve the development of personalized stem cell therapies tailored to each patient's specific needs, the use of gene editing technologies to enhance the therapeutic properties of stem cells, and the exploration of combinatorial approaches to enhance the efficacy of stem cell transplantation [253,254,255].

Stem cell-based therapies have shown potential for treating neurological disorders beyond the well-studied conditions discussed earlier. However, further preclinical and clinical research is needed to establish these approaches' safety, efficacy, and long-term benefits. As regenerative medicine advances, it is hoped that stem cell-based therapies will become a viable treatment option for a wide range of neurological disorders, improving the quality of life for patients and their families.

Conclusion and future perspectives

Stem cell-based therapies for neurological disorders have made significant progress in recent years, with promising results from preclinical studies and early clinical trials. As highlighted in this comprehensive review, various stem cell types, including NSCs, MSCs, and iPSCs, have shown potential for the treatment of a wide range of neurological conditions, such as Parkinson's disease, Alzheimer's disease, multiple sclerosis, stroke, spinal cord injury, and traumatic brain injury.

The therapeutic potential of stem cells in neurological disorders is primarily attributed to their ability to replace lost or damaged neural cells, modulate the immune system, promote endogenous repair mechanisms, and provide trophic support to the injured or diseased nervous system. However, the exact mechanisms underlying the therapeutic effects of stem cells are not fully understood. They may vary depending on the specific neurological condition and the type of stem cells used.

Despite the encouraging progress, several challenges need to be addressed to realize the full potential of stem cell-based therapies for neurological disorders. These include optimizing stem cell sources, differentiation protocols, and delivery methods to ensure the therapeutic approach's safety, efficacy, and reproducibility. The potential for tumorigenicity, immune rejection, and other adverse events following stem cell transplantation also requires careful consideration and long-term monitoring. Furthermore, the complex and multifactorial nature of many neurological disorders may require combinatorial approaches that target multiple pathogenic mechanisms, such as gene therapy, neuroprotective agents, or rehabilitation in conjunction with stem cell transplantation.

Several critical research areas must be prioritized to address these challenges and accelerate the clinical translation of stem cell-based therapies for neurological disorders. These include the development of standardized protocols for the generation, characterization, and banking of clinical-grade stem cells, the establishment of robust preclinical models that more accurately recapitulate human neurological disorders, and the design of well-controlled clinical trials with appropriate patient populations, outcome measures, and follow-up periods.

In addition to technological advancements, the successful clinical translation of stem cell-based therapies for neurological disorders will require a collaborative and multidisciplinary approach involving basic scientists, clinicians, industry partners, regulatory agencies, and patient advocates. Ethical, legal, and social implications of stem cell research and therapy must also be carefully addressed through ongoing dialogue and public engagement.

Looking to the future, the field of stem cell-based therapies for neurological disorders is poised for exciting developments and breakthroughs. The convergence of stem cell biology with other cutting-edge technologies, such as gene editing, single-cell genomics, organoid models, and advanced neuroimaging, holds great promise for developing personalized and targeted therapies for neurological disorders. The increasing understanding of the complex interplay between the nervous system, immune system, and microbiome may also open up new avenues for stem cell-based therapies that harness the body's intrinsic regenerative capacity.

In conclusion, while stem cell-based therapies for neurological disorders are still in their early stages of development, the progress made so far is encouraging and holds great promise for the future. With continued research, collaboration, and innovation, it is hoped that stem cell-based therapies will become a safe, effective, and accessible treatment option for millions worldwide affected by neurological disorders, improving their quality of life and reducing the burden on healthcare systems. As the field continues to evolve, it will be essential to maintain a balanced and evidence-based perspective, acknowledging both the potential and the limitations of stem cell-based therapies and to ensure that the interests of patients and the public are always at the forefront of scientific endeavors.

Data availability

No datasets were generated or analysed during the current study.

References

  1. GBD 2016 Neurology Collaborators. Global, regional, and national burden of neurological disorders, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019;18(5):459–80. https://doi.org/10.1016/S1474-4422(18)30499-X.

    Article  Google Scholar 

  2. Feigin VL, Vos T, Nichols E, et al. The global burden of neurological disorders: translating evidence into policy. Lancet Neurol. 2020;19(3):255–65. https://doi.org/10.1016/S1474-4422(19)30411-9.

    Article  PubMed  Google Scholar 

  3. Thompson AJ, Baranzini SE, Geurts J, Hemmer B, Ciccarelli O. Multiple sclerosis. Lancet. 2018;391(10130):1622–36. https://doi.org/10.1016/S0140-6736(18)30481-1.

    Article  PubMed  Google Scholar 

  4. Ahuja CS, Wilson JR, Nori S, et al. Traumatic spinal cord injury. Nat Rev Dis Primers. 2017;3:17018. https://doi.org/10.1038/nrdp.2017.18.

    Article  PubMed  Google Scholar 

  5. Donnan GA, Fisher M, Macleod M, Davis SM. Stroke. Lancet. 2008;371(9624):1612–23. https://doi.org/10.1016/S0140-6736(08)60694-7.

    Article  CAS  PubMed  Google Scholar 

  6. Maas AIR, Menon DK, Adelson PD, et al. Traumatic brain injury: integrated approaches to improve prevention, clinical care, and research. Lancet Neurol. 2017;16(12):987–1048. https://doi.org/10.1016/S1474-4422(17)30371-X.

    Article  PubMed  Google Scholar 

  7. GBD 2015 Neurological Disorders Collaborator Group. Global, regional, and national burden of neurological disorders during 1990–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet Neurol. 2017;16(11):877–97. https://doi.org/10.1016/S1474-4422(17)30299-5.

    Article  Google Scholar 

  8. Gooch CL, Pracht E, Borenstein AR. The burden of neurological disease in the United States: a summary report and call to action. Ann Neurol. 2017;81(4):479–84. https://doi.org/10.1002/ana.24897.

    Article  PubMed  Google Scholar 

  9. Lang AE, Espay AJ. Disease modification in Parkinson’s disease: current approaches, challenges, and future considerations. Mov Disord. 2018;33(5):660–77.

    Article  PubMed  Google Scholar 

  10. Nichols E, Szoeke CEI, Vollset SE, et al. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019;18(1):88–106. https://doi.org/10.1016/S1474-4422(18)30403-4.

    Article  Google Scholar 

  11. Arthur KC, Calvo A, Price TR, Geiger JT, Chiò A, Traynor BJ. Projected increase in amyotrophic lateral sclerosis from 2015 to 2040. Nat Commun. 2016;7:12408. https://doi.org/10.1038/ncomms12408.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Nagappan PG, Chen H, Wang DY. Neuroregeneration and plasticity: a review of the physiological mechanisms for achieving functional recovery postinjury. Milit Med Res. 2020;7:1–6.

    Google Scholar 

  13. Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer’s disease drug development pipeline: 2020. Alzheimers Dement (N Y). 2020;6(1): e12050. https://doi.org/10.1002/trc2.12050.

    Article  PubMed  Google Scholar 

  14. Wild EJ, Tabrizi SJ. Therapies targeting DNA and RNA in Huntington’s disease. Lancet Neurol. 2017;16(10):837–47. https://doi.org/10.1016/S1474-4422(17)30280-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Res Ther. 2019;10(1):68. https://doi.org/10.1186/s13287-019-1165-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Dulak J, Szade K, Szade A, Nowak W, Józkowicz A. Adult stem cells: hopes and hypes of regenerative medicine. Acta Biochim Pol. 2015;62(3):329–37. https://doi.org/10.18388/abp.2015_1023.

    Article  CAS  PubMed  Google Scholar 

  17. Tabar V, Studer L. Pluripotent stem cells in regenerative medicine: challenges and recent progress. Nat Rev Genet. 2014;15(2):82–92. https://doi.org/10.1038/nrg3563.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13(12):4279–95. https://doi.org/10.1091/mbc.e02-02-0105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Menaa F, Shahrokhi S, Prasad SV. Impact and challenges of mesenchymal stem cells in medicine: an overview of the current knowledge. Stem Cells Int. 2018;2018:5023925. https://doi.org/10.1155/2018/5023925.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Trounson A, McDonald C. Stem cell therapies in clinical trials: progress and challenges. Cell Stem Cell. 2015;17(1):11–22. https://doi.org/10.1016/j.stem.2015.06.007.

    Article  CAS  PubMed  Google Scholar 

  21. Abdi S, Javanmehr N, Ghasemi-Kasman M, Bali HY, Pirzadeh M. Stem cell-based therapeutic and diagnostic approaches in Alzheimer’s disease. Curr Neuropharmacol. 2022;20(6):1093.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Dong XX, Wang Y, Qin ZH. Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative diseases. Acta Pharmacol Sin. 2009;30(4):379–87. https://doi.org/10.1038/aps.2009.24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hosseini SM, Borys B, Karimi-Abdolrezaee S. Neural stem cell therapies for spinal cord injury repair: an update on recent preclinical and clinical advances. Brain. 2024;147(3):766–93.

    Article  PubMed  Google Scholar 

  24. Madhavan L, Daley BF, Paumier KL, Collier TJ. Transplantation of subventricular zone neural precursors induces an endogenous precursor cell response in a rat model of Parkinson’s disease. J Comp Neurol. 2009;515(1):102–15. https://doi.org/10.1002/cne.22033.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Trounson A, DeWitt ND. Pluripotent stem cells progressing to the clinic. Nat Rev Mol Cell Biol. 2016;17(3):194–200. https://doi.org/10.1038/nrm.2016.10.

    Article  CAS  PubMed  Google Scholar 

  26. Lindvall O, Kokaia Z. Stem cells for the treatment of neurological disorders. Nature. 2006;441(7097):1094–6. https://doi.org/10.1038/nature04960.

    Article  CAS  PubMed  Google Scholar 

  27. Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145–7. https://doi.org/10.1126/science.282.5391.1145.

    Article  CAS  PubMed  Google Scholar 

  28. Golchin A, Chatziparasidou A, Ranjbarvan P, Niknam Z, Ardeshirylajimi A. Embryonic stem cells in clinical trials: current overview of developments and challenges. Cell Biology and Translational Medicine, Volume 11: Stem Cell Therapy-Potential and Challenges. 2020 Nov 7:19–37.

  29. Gage FH. Mammalian neural stem cells. Science. 2000;287(5457):1433–8. https://doi.org/10.1126/science.287.5457.1433.

    Article  CAS  PubMed  Google Scholar 

  30. Wu H, Fan Y, Zhang M. Advanced progress in the role of adipose-derived mesenchymal stromal/stem cells in the application of central nervous system disorders. Pharmaceutics. 2023;15(11):2637.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8(9):726–36. https://doi.org/10.1038/nri2395.

    Article  CAS  PubMed  Google Scholar 

  32. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–72. https://doi.org/10.1016/j.cell.2007.11.019.

    Article  CAS  PubMed  Google Scholar 

  33. Shi Y, Inoue H, Wu JC, Yamanaka S. Induced pluripotent stem cell technology: a decade of progress. Nat Rev Drug Discov. 2017;16(2):115–30. https://doi.org/10.1038/nrd.2016.245.

    Article  CAS  PubMed  Google Scholar 

  34. Abbaszadeh H, Ghorbani F, Derakhshani M, Movassaghpour A, Yousefi M. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles: a novel therapeutic paradigm. J Cell Physiol. 2020;235(2):706–17. https://doi.org/10.1002/jcp.29004.

    Article  CAS  PubMed  Google Scholar 

  35. Lindvall O, Kokaia Z. Stem cells in human neurodegenerative disorders–time for clinical translation? J Clin Invest. 2010;120(1):29–40. https://doi.org/10.1172/JCI40543.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Rodríguez-Pallares J, García-Garrote M, Parga JA, Labandeira-García JL. Combined cell-based therapy strategies for the treatment of Parkinson’s disease: focus on mesenchymal stromal cells. Neural Regen Res. 2023;18(3):478–84.

    Article  PubMed  Google Scholar 

  37. Sonntag KC, Song B, Lee N, et al. Pluripotent stem cell-based therapy for Parkinson’s disease: current status and future prospects. Prog Neurobiol. 2018;168:1–20. https://doi.org/10.1016/j.pneurobio.2018.04.005.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Drago D, Cossetti C, Iraci N, et al. The stem cell secretome and its role in brain repair. Biochimie. 2013;95(12):2271–85. https://doi.org/10.1016/j.biochi.2013.06.020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Liang X, Ding Y, Zhang Y, Tse HF, Lian Q. Paracrine mechanisms of mesenchymal stem cell-based therapy: current status and perspectives. Cell Transplant. 2014;23(9):1045–59. https://doi.org/10.3727/096368913X667709.

    Article  PubMed  Google Scholar 

  40. Zhang Y, Chopp M, Meng Y, et al. Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury. J Neurosurg. 2015;122(4):856–67. https://doi.org/10.3171/2014.11.JNS14770.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Xiong Y, Mahmood A, Chopp M. Emerging potential of exosomes for treatment of traumatic brain injury. Neural Regen Res. 2017;12(1):19–22. https://doi.org/10.4103/1673-5374.198966.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Li M, Jiang Y, Hou Q, Zhao Y, Zhong L, Fu X. Potential pre-activation strategies for improving therapeutic efficacy of mesenchymal stem cells: current status and future prospects. Stem Cell Res Ther. 2022;13(1):146.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Zhang W, Xiao D, Mao Q, Xia H. Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther. 2023;8(1):267.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Bátiz LF, Castro MA, Burgos PV, et al. Exosomes as novel regulators of adult neurogenic niches. Front Cell Neurosci. 2016;9:501. https://doi.org/10.3389/fncel.2015.00501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Basmaeil YS, Algudiri D, Alenzi R, et al. Mesenchymal stem cell therapy of Parkinson’s disease: a review of the current status and future perspectives. Stem Cells Int. 2020;2020:5701920. https://doi.org/10.1155/2020/5701920.

    Article  CAS  Google Scholar 

  46. Brummer T, Zipp F, Bittner S. T cell–neuron interaction in inflammatory and progressive multiple sclerosis biology. Curr Opin Neurobiol. 2022;1(75): 102588.

    Article  Google Scholar 

  47. Gugliandolo A, Bramanti P, Mazzon E. Mesenchymal stem cells: a potential therapeutic approach for amyotrophic lateral sclerosis? Stem Cells Int. 2019;2019:3675627. https://doi.org/10.1155/2019/3675627.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Mao AS, Özkale B, Shah NJ, et al. Programmable microencapsulation for enhanced mesenchymal stem cell persistence and immunomodulation. Proc Natl Acad Sci U S A. 2019;116(31):15392–7. https://doi.org/10.1073/pnas.1819415116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gilbert EA, Lakshman N, Lau KS, Morshead CM. Regulating endogenous neural stem cell activation to promote spinal cord injury repair. Cells. 2022;11(5):846.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Han Y, Yang J, Fang J, Zhou Y, Candi E, Wang J, Hua D, Shao C, Shi Y. The secretion profile of mesenchymal stem cells and potential applications in treating human diseases. Signal Transduct Target Ther. 2022;7(1):92.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Geranmayeh MH, Nourazarian A, Avci ÇB, Rahbarghazi R, Farhoudi M. Stem cells as a promising tool for the restoration of brain neurovascular unit and angiogenic orientation. Mol Neurobiol. 2017;54:7689–705.

    Article  CAS  PubMed  Google Scholar 

  52. Hess DC, Wechsler LR, Clark WM, et al. Safety and efficacy of multipotent adult progenitor cells in acute ischaemic stroke (MASTERS): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol. 2017;16(5):360–8. https://doi.org/10.1016/S1474-4422(17)30046-7.

    Article  PubMed  Google Scholar 

  53. Roda AR, Serra-Mir G, Montoliu-Gaya L, Tiessler L, Villegas S. Amyloid-beta peptide and tau protein crosstalk in Alzheimer’s disease. Neural Regen Res. 2022;17(8):1666–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lee AS, Tang C, Rao MS, Weissman IL, Wu JC. Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat Med. 2013;19(8):998–1004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Wood KJ, Issa F, Hester J. Understanding stem cell immunogenicity in therapeutic applications. Trends Immunol. 2016;37(1):5–16.

    Article  CAS  PubMed  Google Scholar 

  56. Lindvall O, Kokaia Z. Stem cell research in stroke: how far from the clinic? Stroke. 2011;42(8):2369–75.

    Article  PubMed  Google Scholar 

  57. Kirchhof K, Feldmann A, Schnabel J, Adler T, Grüschow M, Kögel D, et al. Uncontrolled differentiation is a major risk factor in cell-based therapies for neurodegenerative diseases. Front Cell Dev Biol. 2021;9: 650856.

    Google Scholar 

  58. Lo B, Parham L. Ethical issues in stem cell research. Endocr Rev. 2009;30(3):204–13.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Rivière I, Roy K. Perspectives on manufacturing of high-quality cell therapies. Mol Ther. 2017;25(5):1067–8.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Barker RA, Carpenter MK, Forbes S, Goldman SA, Jamieson C, Murry CE, et al. The challenges of first-in-human stem cell clinical trials: what does this mean for ethics and institutional review boards? Stem Cell Rep. 2018;10(5):1429–31.

    Article  Google Scholar 

  61. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145–7.

    Article  CAS  PubMed  Google Scholar 

  62. Pardo-Moreno T, González-Acedo A, Rivas-Domínguez A, García-Morales V, García-Cozar FJ, Ramos-Rodríguez JJ, Melguizo-Rodríguez L. Therapeutic approach to Alzheimer’s disease: current treatments and new perspectives. Pharmaceutics. 2022;14(6):1117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Zhang G, Li Y, Reuss JL, et al. Stable intracerebral transplantation of neural stem cells for the treatment of paralysis due to ischemic stroke. Stem Cells Transl Med. 2019;8(10):999–1007. https://doi.org/10.1002/sctm.18-0220.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Liang H, Zhao H, Gleichman A, Machnicki M, Telang S, Tang S. Region-specific and activity-dependent regulation of SVZ neurogenesis and recovery after stroke. Proc Natl Acad Sci USA. 2019;116(27):13621–30. https://doi.org/10.1073/pnas.1811825116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Armijo E, Edwards G, Flores A, Vera J, Shahnawaz M, Moda F, Gonzalez C, Sanhueza M, Soto C. Induced pluripotent stem cell-derived neural precursors improve memory, synaptic and pathological abnormalities in a mouse model of Alzheimer’s disease. Cells. 2021;10(7):1802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Skok M. Mesenchymal stem cells as a potential therapeutic tool to cure cognitive impairment caused by neuroinflammation. World J Stem Cells. 2021;13(8):1072.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Liu XY, Yang LP, Zhao L. Stem cell therapy for Alzheimer’s disease. World J Stem Cells. 2020;12(8):787.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Zhao L, Liu JW, Shi HY, Ma YM. Neural stem cell therapy for brain disease. World J Stem Cells. 2021;13(9):1278.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Si Z, Wang X. Stem cell therapies in Alzheimer’s disease: applications for disease modeling. J Pharmacol Exp Ther. 2021;377(2):207–17.

    Article  CAS  PubMed  Google Scholar 

  70. Sun Y, Feng L, Liang L, Stacey GN, Wang C, Wang Y, Hu B. Neuronal cell-based medicines from pluripotent stem cells: development, production, and preclinical assessment. Stem Cells Transl Med. 2021;10(S2):S31-40.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Appelt-Menzel A, Oerter S, Mathew S, Haferkamp U, Hartmann C, Jung M, Neuhaus W, Pless O. Human iPSC-derived blood-brain barrier models: valuable tools for preclinical drug discovery and development? Curr Protoc Stem Cell Biol. 2020;55(1): e122.

    Article  CAS  PubMed  Google Scholar 

  72. Park EH, Lim HS, Lee S, Roh K, Seo KW, Kang KS, Shin K. Intravenous infusion of umbilical cord blood-derived mesenchymal stem cells in rheumatoid arthritis: a phase Ia clinical trial. Stem Cells Transl Med. 2018;7(9):636–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Reis C, Akyol O, Ho WM, et al. Phase I and phase II therapies for acute ischemic stroke: an update on currently studied drugs in clinical research. Biomed Res Int. 2017;2017:4863079. https://doi.org/10.1155/2017/4863079.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Thomsen GM, Gowing G, Svendsen S, Svendsen CN. The past, present and future of stem cell clinical trials for ALS. Exp Neurol. 2014;1(262):127–37.

    Article  Google Scholar 

  75. Zhang Y, Zhang Y, Chopp M, et al. Treatment of traumatic brain injury in rats with N-acetyl-seryl-aspartyl-lysyl-proline. J Neurosurg. 2017;126(3):782–95. https://doi.org/10.3171/2016.3.JNS152699.

    Article  PubMed  Google Scholar 

  76. Cao Z, Kong F, Ding J, Chen C, He F, Deng W. Promoting Alzheimer’s disease research and therapy with stem cell technology. Stem Cell Res Ther. 2024;15(1):1–34.

    Article  CAS  Google Scholar 

  77. Alipour M, Nabavi SM, Arab L, Vosough M, Pakdaman H, Ehsani E, Shahpasand K. Stem cell therapy in Alzheimer’s disease: possible benefits and limiting drawbacks. Mol Biol Rep. 2019;1(46):1425–46.

    Article  Google Scholar 

  78. Novelli G, Spitalieri P, Murdocca M, Centanini E, Sangiuolo F. Organoid factory: the recent role of the human induced pluripotent stem cells (hiPSCs) in precision medicine. Front Cell Dev Biol. 2023;9(10):1059579.

    Article  Google Scholar 

  79. Kumar S, Goyal L, Singh S. Tremor and rigidity in patients with Parkinson’s disease: emphasis on epidemiology, pathophysiology and contributing factors. CNS Neurol Disord Drug Targets. 2022;21(7):596–609.

    Article  CAS  PubMed  Google Scholar 

  80. Cao J, Hou J, Ping J, Cai D. Advances in developing novel therapeutic strategies for Alzheimer’s disease. Mol Neurodegener. 2018;13:1–20.

    Article  Google Scholar 

  81. Heo JY, Nam MH, Yoon HH, Kim J, Hwang YJ, Won W, Woo DH, Lee JA, Park HJ, Jo S, Lee MJ. Aberrant tonic inhibition of dopaminergic neuronal activity causes motor symptoms in animal models of Parkinson’s disease. Curr Biol. 2020;30(2):276–91.

    Article  CAS  PubMed  Google Scholar 

  82. Camilo Jurado-Coronel J, Ávila-Rodriguez M, Capani F, Gonzalez J, Echeverria Moran V, Barreto GE. Targeting the nicotinic acetylcholine receptors (nAChRs) in astrocytes as a potential therapeutic target in Parkinson’s disease. Curr Pharm Design. 2016;22(10):1305–11.

    Article  Google Scholar 

  83. Peng J, Liu Q, Rao MS, Zeng X. Survival and engraftment of dopaminergic neurons manufactured by a Good Manufacturing Practice-compatible process. Cytotherapy. 2014;16(9):1305–12.

    Article  CAS  PubMed  Google Scholar 

  84. Marsili L, Sharma J, Outeiro TF, Colosimo C. Stem cell therapies in movement disorders: lessons from clinical trials. Biomedicines. 2023;11(2):505.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Francis NL, Zhao N, Calvelli HR, Saini A, Gifford JJ, Wagner GC, Cohen RI, Pang ZP, Moghe PV. Peptide-based scaffolds for the culture and transplantation of human dopaminergic neurons. Tissue Eng Part A. 2020;26(3–4):193–205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. 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(13):2133–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Wakeman DR, Hiller BM, Marmion DJ, McMahon CW, Corbett GT, Mangan KP, Ma J, Little LE, Xie Z, Perez-Rosello T, Guzman JN. Cryopreservation maintains functionality of human iPSC dopamine neurons and rescues Parkinsonian phenotypes in vivo. Stem Cell Rep. 2017;9(1):149–61.

    Article  CAS  Google Scholar 

  88. Petrus-Reurer S, Kumar P, Padrell Sánchez S, Aronsson M, André H, Bartuma H, Plaza Reyes A, Nandrot EF, Kvanta A, Lanner F. Preclinical safety studies of human embryonic stem cell-derived retinal pigment epithelial cells for the treatment of age-related macular degeneration. Stem Cells Transl Med. 2020;9(8):936–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Reboussin É, Buffault J, Brignole-Baudouin F, Réaux-Le Goazigo A, Riancho L, Olmiere C, Sahel JA, Mélik Parsadaniantz S, Baudouin C. Evaluation of neuroprotective and immunomodulatory properties of mesenchymal stem cells in an ex vivo retinal explant model. J Neuroinflamm. 2022;19(1):63.

    Article  CAS  Google Scholar 

  90. Shokravi S, Borisov V, Zaman BA, Niazvand F, Hazrati R, Khah MM, Thangavelu L, Marzban S, Sohrabi A, Zamani A. Mesenchymal stromal cells (MSCs) and their exosome in acute liver failure (ALF): a comprehensive review. Stem Cell Res Ther. 2022;13(1):192.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Kordower JH, Vinuela A, Chu Y, Isacson O, Redmond DE Jr. Parkinsonian monkeys with prior levodopa-induced dyskinesias followed by fetal dopamine precursor grafts do not display graft-induced dyskinesias. J Comp Neurol. 2017;525(3):498–512.

    Article  CAS  PubMed  Google Scholar 

  92. Greene PE, Fahn S, Eidelberg D, Bjugstad KB, Breeze RE, Freed CR. Persistent dyskinesias in patients with fetal tissue transplantation for Parkinson disease. Npj Parkinson’s Dis. 2021;7(1):38.

    Article  CAS  Google Scholar 

  93. Christine CW, Richardson RM, Van Laar AD, Thompson ME, Fine EM, Khwaja OS, Li C, Liang GS, Meier A, Roberts EW, Pfau ML. Safety of AADC gene therapy for moderately advanced Parkinson disease: three-year outcomes from the PD-1101 trial. Neurology. 2022;98(1):e40-50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wang YK, Zhu WW, Wu MH, Wu YH, Liu ZX, Liang LM, Sheng C, Hao J, Wang L, Li W, Zhou Q. Human clinical-grade parthenogenetic ESC-derived dopaminergic neurons recover locomotive defects of nonhuman primate models of Parkinson’s disease. Stem Cell Rep. 2018;11(1):171–82.

    Article  CAS  Google Scholar 

  95. 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;12(17):1202027.

    Article  Google Scholar 

  96. Moon H, Kim B, Kwon I, Oh Y. Challenges involved in cell therapy for Parkinson’s disease using human pluripotent stem cells. Front Cell Dev Biol. 2023;11:1288168.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Li Z, Liu F, He X, Yang X, Shan F, Feng J. Exosomes derived from mesenchymal stem cells attenuate inflammation and demyelination of the central nervous system in EAE rats by regulating the polarization of microglia. Int Immunopharmacol. 2019;67:268–80. https://doi.org/10.1016/j.intimp.2018.12.001.

    Article  CAS  PubMed  Google Scholar 

  98. Wang TY, Bruggeman KF, Kauhausen JA, Rodriguez AL, Nisbet DR, Parish CL. Functionalized composite scaffolds improve the engraftment of transplanted dopaminergic progenitors in a mouse model of Parkinson’s disease. Biomaterials. 2016;1(74):89–98.

    Article  Google Scholar 

  99. Flachsbarth K, Jankowiak W, Kruszewski K, Helbing S, Bartsch S, Bartsch U. Pronounced synergistic neuroprotective effect of GDNF and CNTF on axotomized retinal ganglion cells in the adult mouse. Exp Eye Res. 2018;1(176):258–65.

    Article  Google Scholar 

  100. Fymat AL. Multiple sclerosis: I. Symptomatology and etiology. J Neurol Psychol Res. 2023;4:1.

    Google Scholar 

  101. Cadenas-Fernández J, Ahumada-Pascual P, Andreu LS, Velasco A. Recent advances on immunosuppressive drugs and remyelination enhancers for the treatment of multiple sclerosis. Curr Pharm Des. 2021;27(30):3273–80.

    Article  PubMed  Google Scholar 

  102. Yang G, Van Kaer L. Therapeutic targeting of immune cell autophagy in multiple sclerosis: Russian roulette or silver bullet? Front Immunol. 2021;31(12): 724108.

    Article  Google Scholar 

  103. Zveik O, Fainstein N, Rechtman A, Haham N, Ganz T, Lavon I, Brill L, Vaknin-Dembinsky A. Cerebrospinal fluid of progressive multiple sclerosis patients reduces differentiation and immune functions of oligodendrocyte progenitor cells. Glia. 2022;70(6):1191–209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Motavaf M, Sadeghizadeh M, Babashah S, Zare L, Javan M. Dendrosomal nanocurcumin promotes remyelination through induction of oligodendrogenesis in experimental demyelination animal model. J Tissue Eng Regen Med. 2020;14(10):1449–64.

    Article  CAS  PubMed  Google Scholar 

  105. Yao R, Wang B, Ren C, Qu X, Luo M, Zhang Q, Wang H, Dong F, Wu X, Yang L, Yu H. Olig2 overexpression accelerates the differentiation of mouse embryonic stem cells into oligodendrocyte progenitor cells in vitro. Dev Growth Differ. 2014;56(7):511–7.

    Article  CAS  PubMed  Google Scholar 

  106. Sargent A, Shano G, Karl M, Garrison E, Miller C, Miller RH. Transcriptional profiling of mesenchymal stem cells identifies distinct neuroimmune pathways altered by CNS disease. Int J Stem Cells. 2018;11(1):48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Kurte M, Luz-Crawford P, Vega-Letter AM, Contreras RA, Tejedor G, Elizondo-Vega R, Martinez-Viola L, Fernández-O’Ryan C, Figueroa FE, Jorgensen C, Djouad F. IL17/IL17RA as a novel signaling axis driving mesenchymal stem cell therapeutic function in experimental autoimmune encephalomyelitis. Front Immunol. 2018;30(9):802.

    Article  Google Scholar 

  108. Patti F, Chisari CG, Toscano S, Arena S, Finocchiaro C, Cimino V, Milone G. Autologous hematopoietic stem cell transplantation in multiple sclerosis patients: monocentric case series and systematic review of the literature. J Clin Med. 2022;11(4):942.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Mohammadi R, Aryan A, Omrani MD, Ghaderian SM, Fazeli Z. Autologous hematopoietic stem cell transplantation (AHSCT): an evolving treatment avenue in multiple sclerosis. Biol Targets Ther. 2021;15:53–9.

    Article  Google Scholar 

  110. Bose G, Thebault S, Rush CA, Atkins HL, Freedman MS. Autologous hematopoietic stem cell transplantation for multiple sclerosis: a current perspective. Mult Scler J. 2021;27(2):167–73.

    Article  Google Scholar 

  111. Gugliandolo A, Bramanti P, Mazzon E. Mesenchymal stem cells in multiple sclerosis: recent evidence from pre-clinical to clinical studies. Int J Mol Sci. 2020;21(22):8662.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Cohen JA, Imrey PB, Planchon SM, Bermel RA, Fisher E, Fox RJ, Bar-Or A, Sharp SL, Skaramagas TT, Jagodnik P, Karafa M. Pilot trial of intravenous autologous culture-expanded mesenchymal stem cell transplantation in multiple sclerosis. Mult Scler J. 2018;24(4):501–11.

    Article  Google Scholar 

  113. Horak J, Nalos L, Martinkova V, Tegl V, Vistejnova L, Kuncova J, Kohoutova M, Jarkovska D, Dolejsova M, Benes J, Stengl M. Evaluation of mesenchymal stem cell therapy for sepsis: a randomized controlled porcine study. Front Immunol. 2020;7(11):126.

    Article  Google Scholar 

  114. Yuan TF, Dong Y, Zhang L, Qi J, Yao C, Wang Y, Chai R, Liu Y, So KF. Neuromodulation-based stem cell therapy in brain repair: recent advances and future perspectives. Neurosci Bull. 2021;37:735–45.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Wens I, Janssens I, Derdelinckx J, Meena M, Willekens B, Cools N. Made to measure: patient-tailored treatment of multiple sclerosis using cell-based therapies. Int J Mol Sci. 2021;22(14):7536.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Barati S, Tahmasebi F, Faghihi F. Effects of mesenchymal stem cells transplantation on multiple sclerosis patients. Neuropeptides. 2020;1(84): 102095.

    Article  Google Scholar 

  117. Tupone MG, d’Angelo M, Castelli V, Catanesi M, Benedetti E, Cimini A. A state-of-the-art of functional scaffolds for 3D nervous tissue regeneration. Front Bioeng Biotechnol. 2021;18(9): 639765.

    Article  Google Scholar 

  118. Konovalova J, Gerasymchuk D, Parkkinen I, Chmielarz P, Domanskyi A. Interplay between MicroRNAs and oxidative stress in neurodegenerative diseases. Int J Mol Sci. 2019;20(23):6055.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Farina M, Vieira LE, Buttari B, Profumo E, Saso L. The Nrf2 pathway in ischemic stroke: a review. Molecules. 2021;26(16):5001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Kuriakose D, Xiao Z. Pathophysiology and treatment of stroke: present status and future perspectives. Int J Mol Sci. 2020;21(20):7609.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Wu L, Lu J, Lan T, Zhang D, Xu H, Kang Z, Peng F, Wang J. Stem cell therapies: a new era in the treatment of multiple sclerosis. Front Neurol. 2024;9(15):1389697.

    Article  Google Scholar 

  122. Erning K, Segura T. Materials to promote recovery after stroke. Curr Opin Biomed Eng. 2020;1(14):9–17.

    Article  Google Scholar 

  123. Choi YK, Urnukhsaikhan E, Yoon HH, Seo YK, Park JK. Effect of human mesenchymal stem cell transplantation on cerebral ischemic volume-controlled photothrombotic mouse model. Biotechnol J. 2016;11(11):1397–404.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Saffari TM, Bedar M, Hundepool CA, Bishop AT, Shin AY. The role of vascularization in nerve regeneration of nerve graft. Neural Regen Res. 2020;15(9):1573–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Zhai QY, Ren YQ, Ni QS, Song ZH, Ge KL, Guo YL. Transplantation of human umbilical cord mesenchymal stem cells-derived neural stem cells pretreated with Neuregulin1β ameliorate cerebral ischemic reperfusion injury in rats. Biomolecules. 2022;12(3):428.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Nie L, Yao D, Chen S, Wang J, Pan C, Wu D, Liu N, Tang Z. Directional induction of neural stem cells, a new therapy for neurodegenerative diseases and ischemic stroke. Cell Death Discov. 2023;9(1):215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Garcia-Sanchez D, Fernandez D, Rodríguez-Rey JC, Pérez-Campo FM. Enhancing survival, engraftment, and osteogenic potential of mesenchymal stem cells. World J Stem Cells. 2019;11(10):748.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Li Z, Dong X, Tian M, Liu C, Wang K, Li L, Liu Z, Liu J. Stem cell-based therapies for ischemic stroke: a systematic review and meta-analysis of clinical trials. Stem Cell Res Ther. 2020;11:1–3.

    Article  Google Scholar 

  129. Jaillard A, Hommel M, Moisan A, Zeffiro TA, Favre-Wiki IM, Barbieux-Guillot M, Vadot W, Marcel S, Lamalle L, Grand S, Detante O. Autologous mesenchymal stem cells improve motor recovery in subacute ischemic stroke: a randomized clinical trial. Transl Stroke Res. 2020;11:910–23.

    Article  CAS  PubMed  Google Scholar 

  130. Elshaer SL, Bahram SH, Rajashekar P, Gangaraju R, El-Remessy AB. Modulation of mesenchymal stem cells for enhanced therapeutic utility in ischemic vascular diseases. Int J Mol Sci. 2021;23(1):249.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Shafiq M, Jung Y, Kim SH. Insight on stem cell preconditioning and instructive biomaterials to enhance cell adhesion, retention, and engraftment for tissue repair. Biomaterials. 2016;1(90):85–115.

    Article  Google Scholar 

  132. Zera KA, Buckwalter MS. The local and peripheral immune responses to stroke: implications for therapeutic development. Neurotherapeutics. 2020;17(2):414–35.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Weber R. Stem Cell Therapy for Stroke Recovery (Doctoral dissertation, University of Zurich).

  134. Sheikh AM, Hossain S, Tabassum S. Advances in stem cell therapy for stroke: mechanisms, challenges, and future directions. Regen Med Rep. 2024. https://doi.org/10.4103/RMR.REGENMED-D-23-00002.

    Article  Google Scholar 

  135. Pan W, Chen H, Wang A, Wang F, Zhang X. Challenges and strategies: scalable and efficient production of mesenchymal stem cells-derived exosomes for cell-free therapy. Life Sci. 2023;15(319): 121524.

    Article  Google Scholar 

  136. Popa-Wagner A, Sandu RE, Ciobanu O. Co-transplantation Strategies and Combination Therapies for Stroke. Bone marrow stem cell therapy for stroke. 2017:167–200.

  137. Masrori P, Van Damme P. Amyotrophic lateral sclerosis: a clinical review. Eur J Neurol. 2020;27(10):1918–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Kiernan MC, Vucic S, Talbot K, McDermott CJ, Hardiman O, Shefner JM, Al-Chalabi A, Huynh W, Cudkowicz M, Talman P, Van den Berg LH. Improving clinical trial outcomes in amyotrophic lateral sclerosis. Nat Rev Neurol. 2021;17(2):104–18.

    Article  PubMed  Google Scholar 

  139. Mazzini L, Ferrari D, Andjus PR, Buzanska L, Cantello R, De Marchi F, Gelati M, Giniatullin R, Glover JC, Grilli M, Kozlova EN. Advances in stem cell therapy for amyotrophic lateral sclerosis. Expert Opin Biol Ther. 2018;18(8):865–81.

    Article  PubMed  Google Scholar 

  140. Kondo T, Funayama M, Tsukita K, Hotta A, Yasuda A, Nori S, Kaneko S, Nakamura M, Takahashi R, Okano H, Yamanaka S. Focal transplantation of human iPSC-derived glial-rich neural progenitors improves lifespan of ALS mice. Stem Cell Rep. 2014;3(2):242–9.

    Article  CAS  Google Scholar 

  141. Das MM, Avalos P, Suezaki P, Godoy M, Garcia L, Chang CD, Vit JP, Shelley B, Gowing G, Svendsen CN. Human neural progenitors differentiate into astrocytes and protect motor neurons in aging rats. Exp Neurol. 2016;1(280):41–9.

    Article  Google Scholar 

  142. Sironi F, Vallarola A, Violatto MB, Talamini L, Freschi M, De Gioia R, Capelli C, Agostini A, Moscatelli D, Tortarolo M, Bigini P. Multiple intracerebroventricular injections of human umbilical cord mesenchymal stem cells delay motor neurons loss but not disease progression of SOD1G93A mice. Stem Cell Research. 2017;1(25):166–78.

    Article  Google Scholar 

  143. Ciervo Y, Gatto N, Allen C, Grierson A, Ferraiuolo L, Mead RJ, Shaw PJ. Adipose-derived stem cells protect motor neurons and reduce glial activation in both in vitro and in vivo models of ALS. Mol Ther Methods Clin Dev. 2021;11(21):413–33.

    Article  Google Scholar 

  144. Chiarotto GB, Cartarozzi LP, Perez M, Tomiyama AL, de Castro MV, Duarte AS, Luzo ÂC, de Oliveira AL. Delayed onset, immunomodulation, and lifespan improvement of SOD1G93A mice after intravenous injection of human mesenchymal stem cells derived from adipose tissue. Brain Res Bull. 2022;1(186):153–64.

    Article  Google Scholar 

  145. Zhu Q, Lu P. Stem cell transplantation for amyotrophic lateral sclerosis. Stem Cell-Based Ther Neurodegener Dis. 2020:71–97.

  146. Vasques JF, Pinheiro LC, de Jesus Gonçalves RG, Mendez-Otero R, Gubert F. Cell-based research and therapy for amyotrophic lateral sclerosis: promises and challenges. Exon Publications. 2021;25:121–39.

    Google Scholar 

  147. Cho BC, Abreu DR, Hussein M, Cobo M, Patel AJ, Secen N, Lee KH, Massuti B, Hiret S, Yang JC, Barlesi F. Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): primary and follow-up analyses of a randomised, double-blind, phase 2 study. Lancet Oncol. 2022;23(6):781–92.

    Article  CAS  PubMed  Google Scholar 

  148. Je G, Keyhanian K, Ghasemi M. Overview of stem cells therapy in amyotrophic lateral sclerosis. Neurol Res. 2021;43(8):616–32.

    Article  CAS  PubMed  Google Scholar 

  149. Sironi F, De Marchi F, Mazzini L, Bendotti C. Cell therapy in ALS: an update on preclinical and clinical studies. Brain Res Bull. 2023;1(194):64–81.

    Article  Google Scholar 

  150. Liu B, Li M, Zhang L, Chen Z, Lu P. Motor neuron replacement therapy for amyotrophic lateral sclerosis. Neural Regen Res. 2022;17(8):1633–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Reekmans K, Praet J, Daans J, Reumers V, Pauwels P, Van der Linden A, Berneman ZN, Ponsaerts P. Current challenges for the advancement of neural stem cell biology and transplantation research. Stem Cell Rev Rep. 2012;8:262–78.

    Article  PubMed  Google Scholar 

  152. Stanaszek L, Rogujski P, Drela K, Fiedorowicz M, Walczak P, Lukomska B, Janowski M. Transplantation of human glial progenitors to immunodeficient neonatal mice with amyotrophic lateral sclerosis (SOD1/rag2). Antioxidants. 2022;11(6):1050.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Zhao A, Pan Y, Cai S. Patient-specific cells for modeling and decoding amyotrophic lateral sclerosis: advances and challenges. Stem Cell Rev Rep. 2020;16(3):482–502.

    Article  PubMed  Google Scholar 

  154. Sensharma P, Madhumathi G, Jayant RD, Jaiswal AK. Biomaterials and cells for neural tissue engineering: current choices. Mater Sci Eng, C. 2017;1(77):1302–15.

    Article  Google Scholar 

  155. Cheng Y, Zhang Y, Li P. Co-transplantation of autologous treg cells: a groundbreaking cell therapy for brain diseases. J Cereb Blood Flow Meta. 2024. https://doi.org/10.1177/0271678X241245633.

    Article  Google Scholar 

  156. Ng NN, Thakor AS. Locoregional delivery of stem cell–based therapies. Sci Transl Med. 2020;12(547): eaba4564.

    Article  CAS  PubMed  Google Scholar 

  157. Tabrizi SJ, Flower MD, Ross CA, Wild EJ. Huntington disease: new insights into molecular pathogenesis and therapeutic opportunities. Nat Rev Neurol. 2020;16(10):529–46.

    Article  PubMed  Google Scholar 

  158. Eje O, Ogbonna CV, Onoyima CS, Nduka FO. Huntington Disease: mechanism of Pathogenesis and recent developments in its therapeutic strategies: a review. J Chem Rev. 2023;5(2):129–42.

    CAS  Google Scholar 

  159. Palaiogeorgou AM, Papakonstantinou E, Golfinopoulou R, Sigala M, Mitsis T, Papageorgiou L, Diakou I, Pierouli K, Dragoumani K, Spandidos DA, Bacopoulou F. Recent approaches on Huntington’s disease. Biomed Rep. 2023;18(1):1–7.

    Google Scholar 

  160. Islam J, So KH, Kc E, Moon HC, Kim A, Hyun SH, Kim S, Park YS. Transplantation of human embryonic stem cells alleviates motor dysfunction in AAV2-Htt171-82Q transfected rat model of Huntington’s disease. Stem Cell Res Ther. 2021;12:1–4.

    Article  Google Scholar 

  161. Al-Gharaibeh A, Culver R, Stewart AN, Srinageshwar B, Spelde K, Frollo L, Kolli N, Story D, Paladugu L, Anwar S, Crane A. Induced pluripotent stem cell-derived neural stem cell transplantations reduced behavioral deficits and ameliorated neuropathological changes in YAC128 mouse model of Huntington’s disease. Front Neurosci. 2017;10(11):628.

    Article  Google Scholar 

  162. Kim SU, Lee HJ, Kim YB. Neural stem cell-based treatment for neurodegenerative diseases. Neuropathology. 2013;33(5):491–504.

    Article  PubMed  Google Scholar 

  163. Yu-Taeger L, Stricker-Shaver J, Arnold K, Bambynek-Dziuk P, Novati A, Singer E, Lourhmati A, Fabian C, Magg J, Riess O, Schwab M. Intranasal administration of mesenchymal stem cells ameliorates the abnormal dopamine transmission system and inflammatory reaction in the R6/2 mouse model of Huntington disease. Cells. 2019;8(6):595.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Pollock K, Dahlenburg H, Nelson H, Fink KD, Cary W, Hendrix K, Annett G, Torrest A, Deng P, Gutierrez J, Nacey C. Human mesenchymal stem cells genetically engineered to overexpress brain-derived neurotrophic factor improve outcomes in Huntington’s disease mouse models. Mol Ther. 2016;24(5):965–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Shariati A, Nemati R, Sadeghipour Y, Yaghoubi Y, Baghbani R, Javidi K, Zamani M, Hassanzadeh A. Mesenchymal stromal cells (MSCs) for neurodegenerative disease: a promising frontier. Eur J Cell Biol. 2020;99(6): 151097.

    Article  CAS  PubMed  Google Scholar 

  166. Barker RA, Mason SL, Harrower TP, Swain RA, Ho AK, Sahakian BJ, Mathur R, Elneil S, Thornton S, Hurrelbrink C, Armstrong RJ. The long-term safety and efficacy of bilateral transplantation of human fetal striatal tissue in patients with mild to moderate Huntington’s disease. J Neurol Neurosurg Psychiatry. 2013;84(6):657–65.

    Article  PubMed  Google Scholar 

  167. Daviaud N, Garbayo E, Schiller PC, Perez-Pinzon M, Montero-Menei CN. Organotypic cultures as tools for optimizing central nervous system cell therapies. Exp Neurol. 2013;1(248):429–40.

    Article  Google Scholar 

  168. Zimmermann T, Remmers F, Lutz B, Leschik J. ESC-derived BDNF-overexpressing neural progenitors differentially promote recovery in Huntington’s disease models by enhanced striatal differentiation. Stem Cell Rep. 2016;7(4):693–706.

    Article  CAS  Google Scholar 

  169. Benraiss A, Wang S, Herrlinger S, Li X, Chandler-Militello D, Mauceri J, Burm HB, Toner M, Osipovitch M, Jim XuQ, Ding F. Human glia can both induce and rescue aspects of disease phenotype in Huntington disease. Nat Commun. 2016;7(1):11758.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Wu S, FitzGerald KT, Giordano J. On the viability and potential value of stem cells for repair and treatment of central neurotrauma: overview and speculations. Front Neurol. 2018;13(9): 338168.

    Google Scholar 

  171. Caron NS, Dorsey ER, Hayden MR. Therapeutic approaches to Huntington disease: from the bench to the clinic. Nat Rev Drug Discovery. 2018;17(10):729–50.

    Article  CAS  PubMed  Google Scholar 

  172. Cole KL, Findlay MC, Kundu M, Johansen C, Rawanduzy C, Lucke-Wold B. The role of advanced imaging in neurosurgical diagnosis. Journal of modern medical imaging. 2023;1.

  173. Csobonyeiova M, Polak S, Danisovic L. Recent overview of the use of iPSCs Huntington’s disease modeling and therapy. Int J Mol Sci. 2020;21(6):2239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Quadri SA, Farooqui M, Ikram A, Zafar A, Khan MA, Suriya SS, Claus CF, Fiani B, Rahman M, Ramachandran A, Armstrong II. Recent update on basic mechanisms of spinal cord injury. Neurosurg Rev. 2020;43:425–41.

    Article  PubMed  Google Scholar 

  175. Ahuja CS, Nori S, Tetreault L, Wilson J, Kwon B, Harrop J, Choi D, Fehlings MG. Traumatic spinal cord injury—repair and regeneration. Neurosurgery. 2017;80(3S):S9-22.

    Article  PubMed  Google Scholar 

  176. Zipser CM, Cragg JJ, Guest JD, Fehlings MG, Jutzeler CR, Anderson AJ, Curt A. Cell-based and stem-cell-based treatments for spinal cord injury: evidence from clinical trials. Lancet Neurol. 2022;21(7):659–70.

    Article  PubMed  Google Scholar 

  177. Pfister BJ, Gordon T, Loverde JR, Kochar AS, Mackinnon SE, Cullen DK. Biomedical engineering strategies for peripheral nerve repair: surgical applications, state of the art, and future challenges. Crit Rev Biomed Eng. 2011;39(2):81–124. https://doi.org/10.1615/critrevbiomedeng.v39.i2.20.

    Article  PubMed  Google Scholar 

  178. Tomita K, Madura T, Mantovani C, Terenghi G. Differentiated adipose-derived stem cells promote myelination and enhance functional recovery in a rat model of chronic denervation. J Neurosci Res. 2012;90(7):1392–402. https://doi.org/10.1002/jnr.23002.

    Article  CAS  PubMed  Google Scholar 

  179. Chen SY, Yang RL, Wu XC, Zhao DZ, Fu SP, Lin FQ, Li LY, Yu LM, Zhang Q, Zhang T. mesenchymal stem cell transplantation: neuroprotection and nerve regeneration after spinal cord injury. J Inflamm Res. 2023;31:4763–76.

    Article  Google Scholar 

  180. Subramani P, Kannaiyan J, Khare S, Balaji PA, Oyouni AA, Aljohani SAS, Alsulami MO, Al-Amer OM, Alzahrani OR, Altayar MA, Allah Alsulami AA. Toxicity, safety, and efficacy studies on mesenchymal stem cells derived from Decidua basalis in Wistar albino rats by intravenous and subcutaneous routes. Curr Issues Mol Biol. 2022;44(9):4045–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Liau LL, Looi QH, Chia WC, Subramaniam T, Ng MH, Law JX. Treatment of spinal cord injury with mesenchymal stem cells. Cell Biosci. 2020;10:1–7.

    Article  Google Scholar 

  182. Pang QM, Deng KQ, Zhang M, Wu XC, Yang RL, Fu SP, Lin FQ, Zhang Q, Ao J, Zhang T. Multiple strategies enhance the efficacy of MSCs transplantation for spinal cord injury. Biomed Pharmacother. 2023;1(157): 114011.

    Article  Google Scholar 

  183. Xu P, Yang X. The efficacy and safety of mesenchymal stem cell transplantation for spinal cord injury patients: a meta-analysis and systematic review. Cell Transplant. 2019;28(1):36–46.

    Article  PubMed  Google Scholar 

  184. de Araújo LT, Macêdo CT, Damasceno PK, das Neves ÍG, de Lima CS, Santos GC, de Santana TA, Sampaio GL, Silva DN, Villarreal CF, Chaguri AC. Clinical trials using mesenchymal stem cells for spinal cord injury: challenges in generating evidence. Cells. 2022;11(6):1019.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Jin MC, Medress ZA, Azad TD, Doulames VM, Veeravagu A. Stem cell therapies for acute spinal cord injury in humans: a review. Neurosurg Focus. 2019;46(3):E10.

    Article  PubMed  Google Scholar 

  186. Yamazaki K, Kawabori M, Seki T, Houkin K. Clinical trials of stem cell treatment for spinal cord injury. Int J Mol Sci. 2020;21(11):3994.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Zhao X, Li Q, Guo Z, Li Z. Constructing a cell microenvironment with biomaterial scaffolds for stem cell therapy. Stem Cell Res Ther. 2021;12:1–3.

    Article  Google Scholar 

  188. Rocha LA, Silva D, Barata-Antunes S, Cavaleiro H, Gomes ED, Silva NA, Salgado AJ. Cell and tissue instructive materials for central nervous system repair. Adv Func Mater. 2020;30(44):1909083.

    Article  CAS  Google Scholar 

  189. Letko Khait N, Ho E, Shoichet MS. Wielding the double-edged sword of inflammation: building biomaterial-based strategies for immunomodulation in ischemic stroke treatment. Adv Func Mater. 2021;31(44):2010674.

    Article  CAS  Google Scholar 

  190. Davoudi-Monfared E, Abolghasemi R, Allahyari F, Farzanegan G. Adverse events of cell therapy clinical trials in human chronic spinal cord injury, a systematic review and meta-analysis. Regener Ther. 2024;1(27):381–97.

    Article  Google Scholar 

  191. Zeng CW. Multipotent mesenchymal stem cell-based therapies for spinal cord injury: current progress and future prospects. Biology. 2023;12(5):653.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Tashiro S, Nakamura M, Okano H. Regenerative rehabilitation and stem cell therapy targeting chronic spinal cord injury: a review of preclinical studies. Cells. 2022;11(4):685.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Hejrati N, Wong R, Khazaei M, Fehlings MG. How can clinical safety and efficacy concerns in stem cell therapy for spinal cord injury be overcome? Expert Opin Biol Ther. 2023;23(9):883–99.

    Article  CAS  PubMed  Google Scholar 

  194. Ahmed S, Venigalla H, Mekala HM, Dar S, Hassan M, Ayub S. Traumatic brain injury and neuropsychiatric complications. Indian J Psychol Med. 2017;39(2):114–21.

    Article  PubMed  PubMed Central  Google Scholar 

  195. Khatri N, Sumadhura B, Kumar S, Kaundal RK, Sharma S, Datusalia AK. The complexity of secondary cascade consequent to traumatic brain injury: pathobiology and potential treatments. Curr Neuropharmacol. 2021;19(11):1984.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Marklund N, Bellander BM, Godbolt AK, Levin H, McCrory P, Thelin EP. Treatments and rehabilitation in the acute and chronic state of traumatic brain injury. J Intern Med. 2019;285(6):608–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Galgano M, Toshkezi G, Qiu X, Russell T, Chin L, Zhao LR. Traumatic brain injury: current treatment strategies and future endeavors. Cell Transplant. 2017;26(7):1118–30.

    Article  PubMed  PubMed Central  Google Scholar 

  198. Chrostek MR, Fellows EG, Guo WL, Swanson WJ, Crane AT, Cheeran MC, Low WC, Grande AW. Efficacy of cell-based therapies for traumatic brain injuries. Brain Sci. 2019;9(10):270.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Liu S, Shi L, Huang T, Luo Y, Chen Y, Li S, Wang Z. Neural stem cells transplanted into rhesus monkey cortical traumatic brain injury can survive and differentiate into neurons. Int J Mol Sci. 2024;25(3):1642.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Laterza C, Uoshima N, Tornero D, Wilhelmsson U, Stokowska A, Ge R, Pekny M, Lindvall O, Kokaia Z. Attenuation of reactive gliosis in stroke-injured mouse brain does not affect neurogenesis from grafted human iPSC-derived neural progenitors. PLoS ONE. 2018;13(2): e0192118.

    Article  PubMed  PubMed Central  Google Scholar 

  201. Brooks B, Ebedes D, Usmani A, Gonzales-Portillo JV, Gonzales-Portillo D, Borlongan CV. Mesenchymal stromal cells in ischemic brain injury. Cells. 2022;11(6):1013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Guo S, Zhen Y, Wang A. Transplantation of bone mesenchymal stem cells promotes angiogenesis and improves neurological function after traumatic brain injury in mouse. Neuropsychiatr Dis Treat. 2017;6:2757–65.

    Article  Google Scholar 

  203. Yang Y, Ye Y, Su X, He J, Bai W, He X. MSCs-derived exosomes and neuroinflammation, neurogenesis and therapy of traumatic brain injury. Front Cell Neurosci. 2017;28(11):55.

    Google Scholar 

  204. Lengel D, Sevilla C, Romm ZL, Huh JW, Raghupathi R. Stem cell therapy for pediatric traumatic brain injury. Front Neurol. 2020;2(11): 601286.

    Article  Google Scholar 

  205. Cox CS Jr, Notrica DM, Juranek J, Miller JH, Triolo F, Kosmach S, Savitz SI, Adelson PD, Pedroza C, Olson SD, Scott MC. Autologous bone marrow mononuclear cells to treat severe traumatic brain injury in children. Brain. 2024;147(5):1914–25.

    Article  PubMed  Google Scholar 

  206. Wang Z, Luo Y, Chen L, Liang W. Safety of neural stem cell transplantation in patients with severe traumatic brain injury. Exp Ther Med. 2017;13(6):3613–8.

    PubMed  PubMed Central  Google Scholar 

  207. O’Carroll GC, King SL, Carroll S, Perry JL, Vanicek N. The effects of exercise to promote quality of life in individuals with traumatic brain injuries: a systematic review. Brain Inj. 2020;34(13–14):1701–13.

    Article  PubMed  Google Scholar 

  208. Covington NV, Duff MC. Heterogeneity is a hallmark of traumatic brain injury, not a limitation: a new perspective on study design in rehabilitation research. Am J Speech Lang Pathol. 2021;30(2S):974–85.

    Article  PubMed  Google Scholar 

  209. Paik DT, Chandy M, Wu JC. Patient and disease–specific induced pluripotent stem cells for discovery of personalized cardiovascular drugs and therapeutics. Pharmacol Rev. 2020;72(1):320–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Trifirò G, Crisafulli S. A new era of pharmacovigilance: future challenges and opportunities. Front Drug Saf Regul. 2022;25(2):1.

    Google Scholar 

  211. Cozene B, Sadanandan N, Farooq J, Kingsbury C, Park YJ, Wang ZJ, Moscatello A, Saft M, Cho J, Gonzales-Portillo B, Borlongan CV. Mesenchymal stem cell-induced anti-neuroinflammation against traumatic brain injury. Cell Transplant. 2021;23(30):09636897211035715.

    Google Scholar 

  212. Zhang X, Chen X, Hong H, Hu R, Liu J, Liu C. Decellularized extracellular matrix scaffolds: recent trends and emerging strategies in tissue engineering. Bioactive mater. 2022;1(10):15–31.

    Article  Google Scholar 

  213. Zarepour A, Öztürk AB, Irmak DK, Yaşayan G, Gökmen A, Karaöz E, Zarepour A, Zarrabi A, Mostafavi E. Combination therapy using nanomaterials and stem cells to treat spinal cord injuries. Eur J Pharm Biopharm. 2022;1(177):224–40.

    Article  Google Scholar 

  214. Mosilhy EA, Alshial EE, Eltaras MM, Rahman MM, Helmy HI, Elazoul AH, Hamdy O, Mohammed HS. Non-invasive transcranial brain modulation for neurological disorders treatment: a narrative review. Life Sci. 2022;15(307): 120869.

    Article  Google Scholar 

  215. Adiga U, Nandit PB. An overview of pathogenesis of epilepsy. Int Neurourol J. 2023;27(4):88–94.

    Google Scholar 

  216. Johannessen Landmark C, Johannessen SI, Patsalos PN. Therapeutic drug monitoring of antiepileptic drugs: current status and future prospects. Expert Opin Drug Metab Toxicol. 2020;16(3):227–38.

    Article  CAS  PubMed  Google Scholar 

  217. Ching J. Identifying novel therapeutic targets for seizures and brain cancers (Doctoral dissertation, University of Plymouth).

  218. Rosell-Valle C, Martínez-Losa M, Matas-Rico E, Castilla-Ortega E, Zambrana-Infantes E, Gómez-Conde AI, Sánchez-Salido L, Ladrón de Guevara-Miranda D, Pedraza C, Serrano-Castro PJ, Chun J. GABAergic deficits in absence of LPA 1 receptor, associated anxiety-like and coping behaviors, and amelioration by interneuron precursor transplants into the dorsal hippocampus. Brain Struct Funct. 2021;226:1479–95.

    Article  CAS  PubMed  Google Scholar 

  219. Arshad MN, Pinto A, van Praag H, Naegele JR. Altered connectomes of adult-born granule cells following engraftment of GABAergic progenitors in the mouse hippocampus. Prog Neurobiol. 2023;1(226): 102450.

    Article  Google Scholar 

  220. Anderson NC, Van Zandt MA, Shrestha S, Lawrence DB, Gupta J, Chen CY, Harrsch FA, Boyi T, Dundes CE, Aaron G, Naegele JR. Pluripotent stem cell-derived interneuron progenitors mature and restore memory deficits but do not suppress seizures in the epileptic mouse brain. Stem Cell Res. 2018;1(33):83–94.

    Article  Google Scholar 

  221. Aronica E, Bauer S, Bozzi Y, Caleo M, Dingledine R, Gorter JA, Henshall DC, Kaufer D, Koh S, Löscher W, Louboutin JP. Neuroinflammatory targets and treatments for epilepsy validated in experimental models. Epilepsia. 2017;58:27–38.

    Article  PubMed  PubMed Central  Google Scholar 

  222. Salem NA, El-Shamarka M, Khadrawy Y, El-Shebiney S. New prospects of mesenchymal stem cells for ameliorating temporal lobe epilepsy. Inflammopharmacology. 2018;26:963–72.

    Article  CAS  PubMed  Google Scholar 

  223. Alayli A, Lockard G, Gordon J, Connolly J, Monsour M, Schimmel S, Dela Peña I, Borlongan CV. Stem cells: recent developments redefining epilepsy therapy. Cell Transplant. 2023;32:09636897231158967.

    Article  PubMed  PubMed Central  Google Scholar 

  224. Nguyen QT, Thanh LN, Hoang VT, Phan TT, Heke M, Hoang DM. Bone marrow-derived mononuclear cells in the treatment of neurological diseases: knowns and unknowns. Cell Mol Neurobiol. 2023;43(7):3211–50.

    Article  PubMed  Google Scholar 

  225. Milczarek O, Jarocha D, Starowicz-Filip A, Kwiatkowski S, Badyra B, Majka M. Multiple autologous bone marrow-derived CD271+ mesenchymal stem cell transplantation overcomes drug-resistant epilepsy in children. Stem Cells Transl Med. 2018;7(1):20–33.

    Article  CAS  PubMed  Google Scholar 

  226. Ribeiro BF, da Cruz BC, de Sousa BM, Correia PD, David N, Rocha C, Almeida RD, Ribeiro da Cunha M, Marques Baptista AA, Vieira SI. Cell therapies for spinal cord injury: a review of the clinical trials and cell-type therapeutic potential. Brain. 2023;146(7):2672–93.

    Article  PubMed  Google Scholar 

  227. Billakota S, Devinsky O, Kim KW. Why we urgently need improved epilepsy therapies for adult patients. Neuropharmacology. 2020;15(170): 107855.

    Article  Google Scholar 

  228. Mirgh S, Khattry N. Acute complications in stem cell transplantation. Contemporary Bone Marrow Transplantation. 2021:511–44.

  229. Zayed MA, Sultan S, Alsaab HO, Yousof SM, Alrefaei GI, Alsubhi NH, Alkarim S, Al Ghamdi KS, Bagabir SA, Jana A. Stem-cell-based therapy: the celestial weapon against neurological disorders. Cells. 2022;11:3476.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Aquilina K, Chakrapani A, Carr L, Kurian MA, Hargrave D. Convection-enhanced delivery in children: techniques and applications. Adv Tech Stand Neurosurg. 2022;45(18):199–228.

    Article  CAS  PubMed  Google Scholar 

  231. Ramos AG. Enhancing neuronal inhibition by cell and gene therapy as a novel treatment for Epilepsy.

  232. Rahman MM, Islam MR, Islam MT, Harun-Or-Rashid M, Islam M, Abdullah S, Uddin MB, Das S, Rahaman MS, Ahmed M, Alhumaydhi FA. Stem cell transplantation therapy and neurological disorders: current status and future perspectives. Biology. 2022;11(1):147.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Chin EM, Johnson TL, Hoon Jr AH. Cerebral palsy: epidemiology, neurobiology, and lifespan management.

  234. Vitrikas K, Dalton H, Breish D. Cerebral palsy: an overview. Am Fam Physician. 2020;101(4):213–20.

    PubMed  Google Scholar 

  235. Nitkin CR, Rajasingh J, Pisano C, Besner GE, Thébaud B, Sampath V. Stem cell therapy for preventing neonatal diseases in the 21st century: current understanding and challenges. Pediatr Res. 2020;87(2):265–76.

    Article  PubMed  Google Scholar 

  236. Um S, Ha J, Choi SJ, Oh W, Jin HJ. Prospects for the therapeutic development of umbilical cord blood-derived mesenchymal stem cells. World J Stem Cells. 2020;12(12):1511.

    Article  PubMed  PubMed Central  Google Scholar 

  237. Nguyen T, Purcell E, Smith MJ, Penny TR, Paton MC, Zhou L, Jenkin G, Miller SL, McDonald CA, Malhotra A. Umbilical cord blood-derived cell therapy for perinatal brain injury: a systematic review & meta-analysis of preclinical studies. Int J Mol Sci. 2023;24(5):4351.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Lv ZY, Li Y, Liu J. Progress in clinical trials of stem cell therapy for cerebral palsy. Neural Regen Res. 2021;16(7):1377–82.

    Article  PubMed  Google Scholar 

  239. Bhat AN. Motor impairment increases in children with autism spectrum disorder as a function of social communication, cognitive and functional impairment, repetitive behavior severity, and comorbid diagnoses: A SPARK study report. Autism Res. 2021;14(1):202–19.

    Article  PubMed  Google Scholar 

  240. Aishworiya R, Valica T, Hagerman R, Restrepo B. An update on psychopharmacological treatment of autism spectrum disorder. Neurotherapeutics. 2023;19(1):248–62.

    Article  Google Scholar 

  241. Aragón-González A, Shaw PJ, Ferraiuolo L. Blood-brain barrier disruption and its involvement in neurodevelopmental and neurodegenerative disorders. Int J Mol Sci. 2022;23(23):15271.

    Article  PubMed  PubMed Central  Google Scholar 

  242. Guerreiro S, Maciel P. Transition from animal-based to human induced pluripotent stem cells (iPSCs)-based models of neurodevelopmental disorders: opportunities and challenges. Cells. 2023;12(4):538.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Pistollato F, Forbes-Hernández TY, Iglesias RC, Ruiz R, Zabaleta ME, Cianciosi D, Giampieri F, Battino M. Pharmacological, non-pharmacological and stem cell therapies for the management of autism spectrum disorders: a focus on human studies. Pharmacol Res. 2020;1(152): 104579.

    Article  Google Scholar 

  244. Villarreal-Martínez L, González-Martínez G, Sáenz-Flores M, Bautista-Gómez AJ, González-Martínez A, Ortiz-Castillo M, Robles-Sáenz DA, Garza-López E. Stem cell therapy in the treatment of patients with autism spectrum disorder: a systematic review and meta-analysis. Stem Cell Rev Rep. 2022;1:1.

    Google Scholar 

  245. Nasiri M, Parmoon Z, Farahmand Y, Moradi A, Farahmand K, Moradi K, Basti FA, Mohammadi MR, Akhondzadeh S. l-carnitine adjunct to risperidone for treatment of autism spectrum disorder-associated behaviors: a randomized, double-blind clinical trial. Int Clin Psychopharmacol. 2023;7:10–97.

    Google Scholar 

  246. Lopes B, Sousa P, Alvites R, Branquinho M, Sousa AC, Mendonça C, Atayde LM, Luís AL, Varejão AS, Maurício AC. Peripheral nerve injury treatments and advances: one health perspective. Int J Mol Sci. 2022;23(2):918.

    Article  PubMed  PubMed Central  Google Scholar 

  247. de Assis AC, Reis AL, Nunes LV, Ferreira LF, Bilal M, Iqbal HM, Soriano RN. Stem cells and tissue engineering-based therapeutic interventions: promising strategies to improve peripheral nerve regeneration. Cell Mol Neurobiol. 2023;43(2):433–54.

    Article  PubMed  Google Scholar 

  248. Saffari S, Saffari TM, Ulrich DJ, Hovius SE, Shin AY. The interaction of stem cells and vascularity in peripheral nerve regeneration. Neural Regen Res. 2021;16(8):1510–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Saremi J, Mahmoodi N, Rasouli M, Ranjbar FE, Mazaheri EL, Akbari M, Hasanzadeh E, Azami M. Advanced approaches to regenerate spinal cord injury: the development of cell and tissue engineering therapy and combinational treatments. Biomed Pharmacother. 2022;1(146): 112529.

    Article  Google Scholar 

  250. Vijayavenkataraman S. Nerve guide conduits for peripheral nerve injury repair: a review on design, materials and fabrication methods. Acta Biomater. 2020;1(106):54–69.

    Article  Google Scholar 

  251. Lavorato A, Raimondo S, Boido M, Muratori L, Durante G, Cofano F, Vincitorio F, Petrone S, Titolo P, Tartara F, Vercelli A. Mesenchymal stem cell treatment perspectives in peripheral nerve regeneration: systematic review. Int J Mol Sci. 2021;22(2):572.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Diesch-Furlanetto T, Gabriel M, Zajac-Spychala O, Cattoni A, Hoeben BA, Balduzzi A. Late effects after haematopoietic stem cell transplantation in ALL, long-term follow-up and transition: a step into adult life. Front Pediatr. 2021;24(9): 773895.

    Article  Google Scholar 

  253. Aderinto N, Abdulbasit MO, Olatunji D. Stem cell-based combinatorial therapies for spinal cord injury: a narrative review of current research and future directions. Ann Med Surg. 2023;85(8):3943–54.

    Article  Google Scholar 

  254. Li H, Yang Y, Hong W, Huang M, Wu M, Zhao X. Applications of genome editing technology in the targeted therapy of human diseases: mechanisms, advances and prospects. Signal Transduct Target Ther. 2020;5(1):1.

    Article  PubMed  PubMed Central  Google Scholar 

  255. Rahimi Darehbagh R, Mahmoodi M, Amini N, Babahajiani M, Allavaisie A, Moradi Y. The effect of nanomaterials on embryonic stem cell neural differentiation: a systematic review. Eur J Med Res. 2023;28(1):576.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Funding

No financial support has been received from any organization.

Author information

Authors and Affiliations

Authors

Contributions

RRD and NR conceived the idea for the review article. RRD, RR, and SAS performed the literature search and data collection. RRD, SAS, and NR analyzed and interpreted the data. RRD drafted the initial manuscript. All authors critically revised the manuscript for important intellectual content. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Nima Rezaei.

Ethics declarations

Ethics approval and consent participate

This article reviews the existing literature and contains no studies with human participants or animals performed by authors. Therefore, no ethical approval was required for this work.

Declaration of generative AI in scientific writing

While preparing this work, the authors used “Claude 3” AI to check the grammar issues and make the text more narrative. After using this tool, the authors reviewed and edited the content as needed and took full responsibility for the publication's content.

Permission to reproduce material from other sources

Not applicable.

Competing interests

The authors declare no competing interests.

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 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rahimi Darehbagh, R., Seyedoshohadaei, S.A., Ramezani, R. et al. Stem cell therapies for neurological disorders: current progress, challenges, and future perspectives. Eur J Med Res 29, 386 (2024). https://doi.org/10.1186/s40001-024-01987-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-024-01987-1

Keywords