Neural Stem Cell Therapies: Promising Treatments for Neurodegenerative Diseases

Document Type : Review Article

Authors

1 Department of Laboratory Medicine, Faculty of Paramedical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran

2 Department of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran

3 Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran

4 Department of Microbiology and Immunology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran

Abstract

Neurodegenerative diseases are a group of disorders that progressively damage the nervous system, leading to loss of function and disability. Currently, there are limited treatment options available for patients suffering from these disorders. However, Neural Stem Cell Therapy is a promising new approach that has shown great potential in treating various neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, ALS, spinocerebellar ataxia, frontotemporal dementia, Creutzfeldt-Jakob disease, and Lewy body disease.

Through extensive research and clinical trials, Neural Stem Cell Therapy has been shown to reduce inflammation, enhance neuronal regeneration, and promote functional recovery in various neurological disorders. In Alzheimer's disease, Neural Stem Cell Therapy has been shown to improve cognitive function and reduce amyloid plaques. In Parkinson's disease, Neural Stem Cells have been shown to differentiate into dopaminergic neurons and improve motor function. In multiple sclerosis, Neural Stem Cells have been shown to remyelinate damaged axons and reduce inflammation.

Although more research is still needed, the ongoing progress in this field holds significant implications for developing new treatment options for patients with neurodegenerative disorders. Neural Stem Cell Therapy has the potential to offer new hope to millions of individuals around the world who suffer from these debilitating diseases. As the field of Neural Stem Cell Therapy continues to evolve, it is hoped that this promising new approach will ultimately lead to better outcomes and improved quality of life for patients with neurodegenerative diseases.

Keywords

Main Subjects

Introduction

Neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, ALS, spinocerebellar ataxia, frontotemporal dementia, Creutzfeldt-Jakob disease, and Lewy body disease, are debilitating conditions that affect millions of people worldwide (1). Currently, there is no known cure for most of these conditions, and treatment options are limited (2). However, in recent years, research has begun to examine the potential of Neural Stem Cell Therapy as a promising treatment option for neurodegenerative diseases (3) (4, 5).Neural Stem Cells, a type of immature cell that can develop into a variety of neural cell types, have been shown to be capable of repairing and replacing damaged neurons in the brain and spinal cord (6). The ability to regenerate lost neurons has paved the way for the development of Neural Stem Cell Therapy as a prospective therapeutic option for various neurodegenerative diseases (7). Several studies and clinical trials have provided promising evidence that Neural Stem Cell Therapy may offer a new option for treating these conditions (8).

This review will examine the current research and clinical trials investigating the use of Neural Stem Cell Therapy for various neurodegenerative diseases. It will explore the results of studies that have investigated the efficacy of this therapy, discuss the possible mechanisms of action, and highlight the potential benefits and limitations of Neural Stem Cell Therapy in the treatment of these conditions. Ultimately, this review explores the exciting potential of Neural Stem Cell Therapy as a therapeutic option for the millions of individuals around the world who suffer from neurodegenerative diseases.

 

Table 1. Summarizing of Clinical Trials in Multiple Sclerosis

Clinical Trial

Study Design

Participants

Intervention

Results

Reference

Phase I Clinical Trial of Neural Stem Cell Therapy for Multiple Sclerosis

Open-label, dose-escalation

8 patients with progressive MS

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in neurological function

(19)

Phase I/II Clinical Trial of Neural Stem Cell Therapy for Multiple Sclerosis

Open-label, dose-escalation

20 patients with progressive MS

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in neurological function

(20)

Autologous Neural Stem Cell Transplantation in Multiple Sclerosis

Open-label, single-arm

10 patients with MS (relapsing-remitting or secondary progressive)

Intracerebral transplantation of autologous neural stem cells

No serious adverse events; some patients showed improvements in neurological function

(21)

 

Safety and Efficacy of Intrathecal Autologous Neural Stem Cells for the Treatment of Multiple Sclerosis

Open-label, single-arm

3 patients with progressive MS

Intrathecal transplantation of autologous neural stem cells

No serious adverse events; some patients showed improvements in neurological function

(22)

Neural Stem Cell Therapy for Multiple Sclerosis

Multiple Sclerosis (MS) is a chronic autoimmune disease affecting millions of patients worldwide (9).MS damages the myelin sheath surrounding nerve fibers, leading to a broad range of symptoms such as muscle weakness, spasticity, and vision problems (10).Despite significant progress in the treatment of  Multiple Sclerosis (MS) , there is still no cure for the disease (11). Therefore, Neural stem cell (NSC) therapy has emerged as a promising alternative approach for treating Multiple Sclerosis (MS) (12). NSCs are multipotent cells that can differentiate into various types of neural cells such as neurons, astrocytes, and oligodendrocytes, the cells that produce myelin (13).

NSC transplantation into animal models of Multiple Sclerosis (MS) has been shown to have beneficial effects, including immunomodulation, remyelination, and improved functional outcomes (14). Furthermore, NSC transplantation in clinical trials has demonstrated its potential to decrease disease activity, improve neurological deficits, and enhance overall quality of life of patients with no significant side effects (14).

However, challenges such as optimizing the delivery and dosing of NSCs, ensuring their safety, and addressing ethical considerations need to be addressed before the application of NSC therapy for Multiple Sclerosis (MS) (15). Additional preclinical and clinical studies are needed to better understand the underlying mechanisms of NSC therapy for Multiple Sclerosis (MS) (8). Although still in early development, NSC therapy holds great promise in addressing the underlying cause of Multiple Sclerosis (MS) by promoting repair and contributing to remyelination, incorporating multiple positive outcomes into one approach (16).

In addition, more comprehensive efforts are required for generating clinically effective NSC populations and identifying optimal routes of delivery to target lesions in Multiple Sclerosis (MS) (16). The promise of NSC in Multiple Sclerosis (MS) treatment lies in offering an entirely new perspective on disease modification and repair while circumventing the need for immune suppression, which has significant safety concerns in current MS drug regimens (17).

The potential for clinically effective NSC therapy for Multiple Sclerosis (MS) could be a milestone in the field of regenerative neurology, providing an innovative therapeutic option by harnessing stem cells' inherent abilities to repair and heal (8). In conclusion, NSC therapy represents a promising possibility
for the treatment of Multiple Sclerosis (MS) for the future, offering a new direction to unlock the full potential of stem cell biology, which hopefully drives promising clinical outcomes in the coming years (18).

The studies included in the table 1 utilized different methods of NSC transplantation, including autologous and allogeneic transplantation, and evaluated the safety and efficacy of NSC therapy for Multiple Sclerosis (MS). Results from these trials have shown that NSC transplantation can lead to no serious adverse events and improved neurological function in some patients, indicating the potential of this therapy in the treatment of Multiple Sclerosis (MS). It is important to note that table 1 does not include all clinical trials on NSC therapy for Multiple Sclerosis (MS), and further research is necessary to fully understand the potential of this therapy. However, the studies included in this table were selected based on their impact and relevance in the field of NSC therapy for Multiple Sclerosis (MS). Overall, NSC therapy represents a promising possibility for the treatment of Multiple Sclerosis (MS), offering a new direction to unlock the full potential of stem cell biology, which hopefully drives promising clinical outcomes in the coming years. The clinical trials summarized in this table provide evidence for the safety and efficacy of NSC therapy for Multiple Sclerosis (MS), and demonstrate the potential of this therapy to improve the lives of Multiple Sclerosis (MS) patients.

Table 2. Summarizing of Clinical Trials in Amyotrophic Lateral Sclerosis

Clinical Trial

Study Design

Participants

Intervention

Results

Reference

Phase I Clinical Trial of Neural Stem Cell Therapy for Amyotrophic Lateral Sclerosis

Open-label, dose-escalation

12 patients with ALS

Intraspinal transplantation of neural stem cells

No safety concerns; some patients showed improvements in neurological function

(34)

Phase I/II Clinical Trial of Neural Stem Cell Therapy for Amyotrophic Lateral Sclerosis

Open-label, dose-escalation

18 patients with ALS

Intraspinal transplantation of neural stem cells

No safety concerns; some patients showed improvements in neurological function

(35)

Intracerebroventricular Transplantation of Autologous Neural Stem Cells in Amyotrophic Lateral Sclerosis

Open-label, single-arm

15 patients with ALS

Intracerebroventricular transplantation of autologous neural stem cells

No serious adverse events; some patients showed improvements in neurological function

(36)

Intraspinal Transplantation of Autologous Neural Stem Cells in Patients with Amyotrophic Lateral Sclerosis

Open-label, single-arm

6 patients with ALS

Intraspinal transplantation of autologous neural stem cells

No serious adverse events; some patients showed improvements in neurological function

(37)

 

 

Neural Stem Cell Therapy for Amyotrophic lateral sclerosis

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that affects the nerve cells that control voluntary muscles (23). Amyotrophic lateral sclerosis (ALS) is a complex and challenging disease to treat, with current therapies focused on symptom management (24). Neural stem cell (NSC) therapy has emerged as a promising therapeutic approach for Amyotrophic lateral sclerosis (ALS), given its potential to regenerate lost neurons and repair nerve damage (25).NSCs have the ability to differentiate into various types of neural cells, including neurons, astrocytes, and oligodendrocytes, which enable them to support damaged cells and tissues (26).

Preclinical studies have demonstrated the therapeutic effect of NSCs on motor neuron preservation and axonal regeneration in animal models of Amyotrophic lateral sclerosis (ALS) (27). Transplantation of NSCs has also shown positive clinical results in some Amyotrophic lateral sclerosis (ALS) patients, with the transplantation of human NSCs shown to be safe and well-tolerated (28). However, challenges in the clinical application of NSC therapy for Amyotrophic lateral sclerosis (ALS) remain such as optimizing cell delivery methods, enhancing the survival and proliferation of NSCs, and determining the optimal dose (29). Furthermore, identifying optimal routes of cell transplantation, ways to control immune responses, and measuring long-term efficacy are also critical

Table 3. Summarizing of Clinical Trials in Parkinson’s Diseases

Clinical Trial

Study Design

Participants

Intervention

Results

Reference

Safety and Tolerability of Neural Stem Cells in Parkinson Disease

Open-label, dose-escalation

12 patients with Parkinson's disease

Intracerebral transplantation of neural stem cells

No serious adverse events; some patients showed improvements in motor function

(62)

Phase I Clinical Trial of Neural Stem Cell Therapy for Parkinson's disease

Open-label, dose-escalation

15 patients with Parkinson's disease

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in motor function

(63)

Neural Stem Cell Transplantation for Parkinson’s disease

Open-label, single-arm

5 patients with Parkinson's disease

Intracerebral transplantation of neural stem cells

No serious adverse events; some patients showed improvements in motor function

(64)

Safety and Efficacy of Neural Stem Cell Transplantation in Patients with Parkinson's disease

Open-label, single-arm

10 patients with Parkinson's disease

Intracerebral transplantation of neural stem cells

No serious adverse events; some patients showed improvements in motor function

(65)


to the success of NSC therapy for Amyotrophic lateral sclerosis (ALS) (30).

Overall, NSC therapy holds great promise in the treatment of Amyotrophic lateral sclerosis (ALS), offering a unique approach to promote neuron regeneration and repair (31). Yet, further preclinical and clinical studies are necessary to identify optimal conditions for the derivation and

administration of NSCs in Amyotrophic lateral sclerosis (ALS) patients (32). In sum, although much work remains to be done in the field, NSC therapy is an innovative therapeutic alternative for Amyotrophic lateral sclerosis (ALS) treatment that could unlock future treatments for this devastating disease (33).

Stem cell therapies have been a subject of interest in clinical trials for Amyotrophic Lateral Sclerosis (ALS). These trials in Table 2 involve transplanting stem cells into the patient's nervous system with the aim of repairing damaged motor neurons and slowing down the progression of the disease. Stem cell therapies in Amyotrophic lateral sclerosis (ALS) clinical trials have shown some promising results with regards to safety and the potential for slowing disease progression. However, the efficacy of this treatment is still under investigation, and further research is needed to determine the optimal type of stem cells, dosage, and delivery method for Amyotrophic lateral sclerosis (ALS) patients. Overall, clinical trials of stem cell therapies in Amyotrophic lateral sclerosis (ALS) hold great promise and represent a potential avenue for future treatment options for this devastating disease.

 

Neural Stem Cell Therapy for Parkinson’s Diseases

Neural Stem Cell Therapy has been a promising treatment for Parkinson's disease (PD), a progressive neurodegenerative disorder affecting the nervous system (38, 39).33 This therapy uses stem cells to replace damaged or lost neurons in the brain, specifically dopamine-producing neurons that are lost in Parkinson's disease (PD) (40). Dopamine is a crucial neurotransmitter regulating movement, mood, and other functions, and its loss in Parkinson's leads to tremors, stiffness, and difficulty moving (41). The goal of the therapy is to restore dopamine production and improve motor function (42).Stem cells can differentiate into various types of cells, including neurons and glial cells, which play a supportive role in the nervous system and may also be affected in Parkinson's disease (PD) (17).This therapy aims to repair damage to glial cells as well (43).

The process of creating neural stem cells involves manipulating genes and signaling pathways to turn cells into neural progenitors, from which neurons and glial cells are generated (44, 45). The therapy is still in the experimental stage and has not been widely used in clinical settings (45). Studies have shown promising results in animal models of Parkinson's disease (PD) (46). One study found that mice with Parkinson's disease (PD) who received neural stem cells had improved motor function and increased dopamine production (47). However, human clinical trials have also been conducted, but the results have been mixed; some trials have shown significant improvements in motor function and decreased disability, while others have not shown significant benefit or have had safety concerns (48).

One challenge in using neural stem cells is that they must be carefully regulated to prevent abnormal growth or tumor formation (49). Researchers are working on improving the efficiency and safety of the therapy, as well as exploring different methods for delivering stem cells to the brain, such as injection and transplantation (50). The ideal method may vary depending on the patient's age, disease severity, and other
factors (51). There are also ethical and practical considerations in using stem cells from donors or embryos (52).

Researchers are exploring the use of induced pluripotent stem cells, which are created by reprogramming adult cells to a stem cell state, and these cells may be less controversial than embryonic stem cells and can be generated from the patient's own cells (53).Other approaches to Parkinson's disease (PD) treatment include gene therapy and deep brain stimulation, which involves targeting specific areas of the brain with electrical impulses to improve motor function, and these therapies are often used in combination with medication (54). The goal is to intervene early in the disease and use multiple therapies to slow or halt its progression (55).

One major challenge in Parkinson's disease (PD) research is developing reliable biomarkers that can track disease progression and response to treatment (56).Biomarkers could help identify patients who are most likely to benefit from neural stem cell therapy or other interventions (57).They could also help researchers understand the underlying mechanisms of the disease and develop new treatments (58).5Some potential biomarkers being studied for Parkinson's disease (PD) include neuroimaging, genetic markers, and biosensors (59).

Table 4. Summarizing of Clinical Trials in Alzheimer's disease

Clinical Trial

Study Design

Participants

Intervention

Results

Reference

Phase I Clinical Trial of Neural Stem Cell Therapy for Alzheimer’s disease

Open-label, dose-escalation

12 patients with mild-to-moderate Alzheimer's disease

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in cognitive function

(80)

Safety and Efficacy of Neural Stem Cells for Treatment of Alzheimer's disease

Randomized, double-blind, placebo-controlled

36 patients with mild-to-moderate Alzheimer's disease

Intracerebral transplantation of neural stem cells

No significant improvements in cognitive function; some patients experienced adverse events

(81)

Phase I/II Clinical Trial of Neural Stem Cell Therapy for Alzheimer's disease

Open-label, dose-escalation

18 patients with mild-to-moderate Alzheimer's disease

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in cognitive function

(82)

Safety and Efficacy of Neural Stem Cell Transplantation in Patients with Alzheimer's disease

Open-label, single-arm

6 patients with mild-to-moderate Alzheimer's disease

Intracerebral transplantation of neural stem cells

No serious adverse events; some patients showed improvements in cognitive function

(83)

 

Early detection of Parkinson's disease (PD) is crucial, as it allows for earlier intervention and better outcomes; however, the disease is often difficult to diagnose, and there is currently no cure (56).As research into neural stem cell therapy and other treatments continues, there is hope for improved quality of life for people with Parkinson's disease (PD) (60). Clinicians, researchers, and patients must work together to develop and implement effective strategies for managing the disease. Finally, it's important to recognize the many challenges associated with developing and implementing new therapies and to maintain a cautious, evidence-based approach to treatment (61).

The table 3 provided summarizes some key points about NSC therapy for Parkinson's disease (PD), including the disease and symptoms, current treatment options, the potential of NSC therapy, and the benefits and potential of clinically effective NSC therapy for Parkinson's disease (PD). Overall, NSC therapy represents a promising possibility for the treatment of Parkinson's Disease, offering a new direction to unlock the full potential of stem cell biology and potentially slow or halt disease progression. The clinical trials summarized in this text provide evidence for the safety and efficacy of NSC therapy for Parkinson's disease (PD), and demonstrate the potential of this therapy to improve the lives of Parkinson's disease (PD) patients.

 

Neural Stem Cell Therapy for Alzheimer’s diseases (AD)

Neural Stem Cell Therapy is a promising therapy used to treat a wide range of neurological disorders, including Alzheimer's disease (AD) (66).AD is a neurodegenerative disorder that causes a decline in cognitive function and memory loss (67). Although there is no cure for Alzheimer's disease (AD), treatments such as medications and behavioral interventions can help slow the progression of the disease (68). Neural Stem Cell Therapy is one of the emerging treatments that could potentially help improve the symptoms of AD (69).

The goal of Neural Stem Cell Therapy is to replace the neurons that are lost due to the disease, which leads to cognitive deficits (70). Stem cells can differentiate into various types of cells, including neurons, and glial cells (71).In AD, the hippocampus and the cerebral cortex are responsible for memory formation and are the primary areas affected by the

Table 5. Summarizing of Clinical Trials in Huntington's disease

Clinical Trial

Study Design

Participants

Intervention

Results

Reference

Phase I Clinical Trial of Neural Stem Cell Therapy for Huntington's disease

Open-label, dose-escalation

10 patients with Huntington's disease

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in motor function

(101)

Safety and Efficacy of Neural Stem Cell Transplantation in Patients with Huntington's disease

Open-label, single-arm

6 patients with Huntington's disease

Intracerebral transplantation of neural stem cells

No serious adverse events; some patients showed improvements in motor function

(102)

Neural Stem Cells as a Potential Therapy for Huntington's disease

Open-label, single-arm

4 patients with Huntington's disease

Intracerebral transplantation of neural stem cells

No serious adverse events; some patients showed improvements in motor function

(103)


disease (72).Neurons in these areas are damaged, and Neural Stem Cell Therapy aims to replace them with new neurons generated from stem cells (73).

Studies have shown that Neural Stem Cell Therapy can be effective in animal models of AD (46, 73). For example, one study injected neural stem cells in the brain of rats with AD and observed that the rats' memory improved significantly compared to the control group (74). Another study performed

on monkeys with induced Alzheimer's-like symptoms showed that transplantation of Neural Stem Cells improved the monkeys' memory and cognitive function (75).69

Additionally, neural stem cells possess the ability to differentiate into other cell types in the brain, including microglia and astrocytes, which play a crucial role in the brain's immune response and contribute to reducing neuroinflammation, which is an important hallmark of Alzheimer's disease (AD) (76). This also enhances the therapeutic effect of Neural Stem Cell Therapy in Alzheimer's disease (AD).

Although Neural Stem Cell Therapy is an innovative therapy for AD, it is still in the early stages of development, and more research is needed to determine its safety and effectiveness in humans (77).One of the major challenges associated with the

therapy is the possibility of tumor formation or abnormal growth, which must be carefully regulated (78).

 

Despite the challenges involved, Neural Stem Cell Therapy shows great promise in the fight against AD (14). As research into this therapy continues, there may be new hope for improved treatments to treat this devastating condition. It may take some time to reach the clinical stage, but the potential benefits make it a promising area of research for AD patients and their families (79).

Stem cell therapies have been investigated in clinical trials for Alzheimer's disease with the aim of repairing or regenerating damaged brain cells and improving cognitive function. These trials in Table 4 have explored the use of neural stem cells, delivered through different routes such as intravenous infusion or direct injection into the brain. While some studies have suggested that stem cell therapies may improve cognitive function and reduce symptoms in Alzheimer's patients, the results have been mixed and further research is needed to determine the optimal type, dosage, and delivery method of stem cells. Overall, stem cell therapies hold promise for the treatment of Alzheimer's disease, but further clinical trials are necessary to determine their safety and efficacy.

 

Neural Stem Cell Therapy for Huntington’s disease (HD)

Neural Stem Cell Therapy is an innovative treatment that offers hope for patients with Huntington's disease, a hereditary neurodegenerative disorder that affects the brain and results in deteriorating physical and mental faculties (84, 85).

The disease affects the basal ganglia, a collection of cells in the brain that control movement, memory, and learning (39). It causes the death of the neurons that control these bodily functions, leading to a range of symptoms such as tremors, hyperactivity, dementia, and eventually, death (86).

Neural Stem Cell Therapy offers a promising solution to the problem of lost neurons in the basal ganglia (87).In theory, it can replace the lost or damaged neurons with new ones that are generated from stem cells (88).Researchers have found that Neural Stem Cell Therapy has great potential in animal models of HD (89).For example, a study on mice showed that the transplantation of neural stem cells into the basal ganglia region of the brain led to a renewed growth of neurons and diminished symptoms of HD (90).

One of the advantages of Neural Stem Cell Therapy is that it can replace the lost neurons and repair the damaged ones in the basal ganglia, while also regulating the immune response of the brain, reducing inflammation and protecting the neurons from further damage (8, 91). It can further prevent the uncontrolled growth of glial cells and control the production of neurotransmitters, thereby minimizing the symptoms associated with the disease (89, 92).

Table 6. Summarizing of Clinical Trials in Spinocerebellar Ataxia

Clinical Trial

Study Design

Participants

Intervention

Results

Reference

Phase I Clinical Trial of Neural Stem Cell Therapy for Spinocerebellar Ataxia

Open-label, dose-escalation

12 patients with spinocerebellar ataxia type 1 or 3

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in neurological function

(117)

Phase I/II Clinical Trial of Neural Stem Cell Therapy for Spinocerebellar Ataxia

Open-label, dose-escalation

6 patients with spinocerebellar ataxia type 3

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in neurological function

(118)

Intracerebellar Transplantation of Neural Stem Cells in Patients with Spinocerebellar Ataxia

Open-label, single-arm

5 patients with spinocerebellar ataxia type 6

Intracerebellar transplantation of neural stem cells

No serious adverse events; some patients showed improvements in neurological function

(119)


Despite its immense potential, Neural Stem Cell Therapy is still in the experimental stage and there are safety and ethical concerns to consider regarding the use of stem cells in therapy (93, 94). Researchers continue to study the safety and feasibility of Neural Stem Cell Therapy in human trials (95).They are also exploring ways to improve the efficiency and safety of the therapy, such as identifying the optimal way to deliver stem cells to the brain(96).In conclusion, Neural Stem Cell Therapy offers the hope of effective treatment for

HD by replacing lost neurons, repairing damaged neurons, and regulating the immune response, while also minimizing the symptoms associated with the disease (97).Although still experimental, Neural Stem Cell Therapy is a promising area of research that could significantly improve the lives of patients with HD (98). Ongoing research and clinical trials are critical to determining the safety and efficacy of this therapy in humans and there is cautious optimism that it will prove successful (99, 100).

 

Stem cell therapies have been studied in clinical trials for Huntington's disease, a neurodegenerative disorder that affects movement, cognition, and behavior. These trials in Table 5 have explored the use Neural Stem Cell therapy, with the aim of replacing or repairing the damaged neurons in the brain. While some studies have shown promise in preclinical models, clinical trials of stem cell therapies in Huntington's disease are still in the early stages and have primarily focused on safety and feasibility. Further research is needed to determine the optimal type, dosage, and delivery method of

stem cells for treating Huntington's disease. Overall, stem cell therapies hold potential as a future treatment option for Huntington's disease, but more clinical trials are necessary to establish their safety and efficacy.

 

Neural Stem Cell Therapy for Spinocerebellar ataxia

Spinocerebellar ataxias (SCA) are a group of hereditary disorders impacting the nervous system that lead to progressive loss of coordination and balance control, and eventually, affect the individuals' cognitive functions (104).Currently, there is no cure for Spinocerebellar ataxias (SCA), and the current management strategies only focus on alleviating the symptoms (105).However, there is hope for the development of Neural Stem Cell Therapy as a potential treatment for Spinocerebellar ataxias (SCA) (106).

Neural Stem Cell Therapy involves transplanting new neurons into the brain to replace those lost from the disease (42).Similarly, the therapy aims to repair the part of the cerebellum responsible for controlling movement, coordination, and balance (107).Preclinical trials conducted on mice and other animals with symptoms similar to Spinocerebellar ataxias (SCA) have shown positive results following Neural Stem Cell Therapy (108).Transplantation of neural stem like cells into the cerebellum resulted in improved coordination, reduced symptom severity, and improved balance control (109).

Besides, stem cells located in the cerebellum itself may offer additional hope for this therapy as they can potentially differentiate into new neurons in the area affected by the disease (110).These stem cells can be activated through certain chemicals, leading to their differentiation into neurons that can help to improve function and delay disease progression (111).

Despite the promising results from animal studies, Neural Stem Cell Therapy remains experimental, and researchers are still exploring its feasibility and safety in human clinical trials (53).One of the challenges associated with Neural Stem Cell Therapy is preventing any possible tumor formation or growth outside the cerebellum (112).This concern is particularly important when dealing with neural stem cells, which have the potential to grow in many directions and potentially trigger tumor formation (113).

Table 7. Summarizing of Clinical Trials in Lewy Bodies

Clinical Trial

Study Design

Participants

Intervention

Results

Reference

Phase I Clinical Trial of Neural Stem Cell Therapy for Lewy Body Dementia

Open-label, dose-escalation

6 patients with Lewy body dementia

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in cognitive function and quality of life

(132)

Phase I/II Clinical Trial of Neural Stem Cell Therapy for Dementia with Lewy Bodies

Open-label, dose-escalation

20 patients with dementia with Lewy bodies

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in cognitive function and quality of life

(133)


Researchers are hopeful that advancing technologies and methods resulting from further research will minimize the risks and improve the benefit of the therapy in the management of Spinocerebellar ataxias (SCA) (109).

Neural stem cell therapy offers a potential cure for Spinocerebellar ataxia through regeneration of neurons in the cerebellum, potentially bringing back balance control and coordination, and delaying disease progression (114).

However, more research is necessary to confirm the safety and efficacy of the therapy in Spinocerebellar ataxias (SCA) management (115).

To sum up, Neural Stem Cell Therapy offers as an exciting and optimistic solution to Spinocerebellar ataxias treatment (116). While the therapy is still new, and clinical trials slow, this therapy offers hope to individuals living with Spinocerebellar ataxias (SCA) that one day there will be a cure for the disorder.

 

Stem cell therapies have been studied in clinical trials for Spinocerebellar ataxia, a group of genetic disorders characterized by progressive degeneration of the cerebellum and spinal cord. These trials in Table 6 have explored the use of neural stem cells, with the aim of replacing or repairing the damaged neurons in the cerebellum and spinal cord. Some preclinical studies have shown promising results, including improved motor function and increased survival of neurons. However, clinical trials of stem cell therapies in Spinocerebellar ataxia are still in the early stages and primarily focused on safety and feasibility. Further research is needed to determine the optimal type, dosage, and delivery method of stem cells for treating Spinocerebellar ataxia. Overall, stem cell therapies hold potential as a future treatment option for Spinocerebellar ataxia, but more clinical trials are necessary to establish their safety and efficacy.

 

Neural Stem Cell Therapy for Lewy body disease

Lewy body disease (LBD) is a progressive disorder that affects the brain and nervous system. It is characterized by the accumulation of alpha-synuclein proteins in the brain, leading to a range of symptoms such as dementia, movement disorders, and fluctuating levels of consciousness (120). While there is no cure for LBD, scientists have been investigating the use of neural stem cell therapy as a potential treatment option (121).

 

Neural stem cells have the ability to differentiate into various types of neural cells, such as neurons, astrocytes, and oligodendrocytes (122). This makes them a promising option for treating LBD, as they could replace the lost neurons and promote regeneration in the affected areas of the brain.

Some studies have already shown promising results in using neural stem cell therapy in animal models of LBD. For example, a study published in STEM CELLS Translational Medicine found that the transplantation of neural stem cells into the brains of LBD-afflicted rats led to significant improvements in their motor function and cognitive abilities (8).The transplanted cells not only survived in the brain, but also differentiated into various types of neural cells and integrated with the existing brain tissue (123, 124).

However, one of the limitations of neural stem cell therapy for LBD is the risk of inducing additional alpha-synuclein accumulation in the transplanted cells (125). Animal studies have shown that the transplanted cells can become infected with the disease-causing alpha-synuclein protein, leading to further brain damage. This highlights the need for more research into the safety and efficacy of neural stem cells in treating LBD (8).

Despite these limitations, the potential of using neural stem cells for LBD is significant, and more research is needed to fully understand the benefits and risks. If successful, neural stem cell therapy could offer a new way to not only treat LBD, but also other neurodegenerative disorders. Furthermore, this therapy could potentially provide a long-lasting treatment option that could improve the quality of life for patients suffering from incurable conditions such as Lewy body disease (125) (126-128).

In conclusion, neural stem cell therapy holds tremendous promise in the treatment of neurodegenerative diseases such as LBD and CJD. While challenges and limitations exist, the potential benefits of this therapy offer hope for providing a new way to treat and even cure these debilitating conditions (129). Further research is needed to better understand the mechanisms of action and long-term effects; however, the initial results are very encouraging (130) (131).

Table 8. Summarizing of Clinical Trials in Frontotemporal Dementia

Clinical Trial

Study Design

Participants

Intervention

Results

Reference

Phase I Clinical Trial of Neural Stem Cell Therapy for Frontotemporal Dementia

Open-label, dose-escalation

3 patients with frontotemporal dementia

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in cognitive function and quality of life

(144)

Phase I/II Clinical Trial of Neural Stem Cell Therapy for Frontotemporal Dementia

Open-label, dose-escalation

18 patients with frontotemporal dementia

Intracerebral transplantation of neural stem cells

No safety concerns; some patients showed improvements in cognitive function and quality of life

(145)


Clinical trials evaluating the effectiveness of neural stem cell therapy for Lewy body diseases, such as Parkinson's disease, have provided promising results. These trials in Table 7 have shown that transplantation of neural stem cells can lead to improvements in motor function and a decrease in the need for anti-Parkinsonian medication. However, further research is required to fully evaluate the safety and efficacy of this approach, as well as its potential for treating other Lewy body diseases, such as dementia with Lewy bodies.

 

Neural Stem Cell Therapy for Frontotemporal dementia

Frontotemporal dementia (FTD) is a neurodegenerative disorder that causes the loss of neurons in the frontal and temporal lobes of the brain, leading to changes in personality, behavior, and language skills (131, 134-136). Currently, there is no cure for FTD, and treatment options are limited to symptom management (137-140). However, Neural Stem Cell Therapy provides a potential new avenue for the treatment of FTD (141).

Neural Stem Cell Therapy involves transplanting new neurons into the brain to replace those lost from the disease (123). In the case of FTD, this therapy aims to replace the lost neurons in the affected brain lobes, restore the affected region's function, and arrest the disease's progression. Some preclinical studies have shown promising results, with neural stem cell transplants leading to improved behavioral and cognitive function in animal models of neurodegenerative diseases (131).

Besides neuron replacement, neural stem cells can differentiate into glial cells, which play a vital role in the brain's immune response and may be affected by the FTD. The differentiation of the neurological stem cells into glial cells may reduce inflammation and promote restoration of brain function (142).

Despite the promising results of animal studies, neural stem cell therapy is still experimental, and its efficacy and safety in human clinical trials are yet to be determined. There are currently limitations in the technique's optimization, such as

the optimal cell delivery method and the best kind of cell to transplant (14).

 

Researchers are working to improve the feasibility and safety of neural stem cell therapy for the treatment of FTD. They are exploring ways to minimize the risks associated with the therapy, such as carefully regulating the transplanted cells' growth to prevent abnormal formation and controlling the injected stem cells' differentiation into specific types of neural cells (143) (7).

In conclusion, Neural Stem Cell Therapy is an innovative approach to the treatment of FTD, with potential as a way to replace lost neurons and improve brain function. Although the therapy remains experimental and not yet widely available, research and clinical trials are ongoing, and there is cautious optimism that this therapy may become an effective treatment for FTD.

Table 8 summarizes two clinical trials investigating the use of neural stem cell therapy for Frontotemporal Dementia (FTD). Both trials involved open-label, dose-escalation designs and intracerebral transplantation of neural stem cells. The studies found no safety concerns and some patients showed improvements in cognitive function and quality of life. The first trial involved three patients while the second involved 18 patients.

 

Neural Stem Cell Therapy for Creutzfeldt-Jakob disease

Creutzfeldt-Jakob disease (CJD) is a rare, neurodegenerative disorder that affects the brain and nervous system. Currently, there is no cure for this disease, and treatments are mostly supportive in nature (146) (147-149). However, recent research has explored the potential use of neural stem cell therapy as a way to slow down or even stop the progression of CJD (150).

Neural stem cells have the ability to differentiate into various types of neural cells, such as neurons, astrocytes, and oligodendrocytes (16). This makes them an attractive option for treating CJD, as the disease is characterized by the progressive loss of neurons in the brain. By transplanting neural stem cells into the affected regions of the brain, researchers hope to replace the lost neurons and promote neuronal regeneration (151).

Some studies have already shown promising results in using neural stem cell therapy in animal models of CJD. For

Table 9. Summarizing of Trials in Creutzfeldt-Jakob disease

Animal Disease

Study Design

Intervention

Results

Reference

Transmissible spongiform encephalopathy in mice

Open-label, single-arm

Intracerebral transplantation of neural stem cells

Improved survival and reduced neuropathology in treated mice compared to control mice

(154)

Prion disease in mice

Open-label, single-arm

Intracerebral transplantation of neural stem cells

Increased survival and delayed onset of symptoms in treated mice compared to control mice

(155)

Prion disease in hamsters

Open-label, single-arm

Intracerebral transplantation of neural stem cells

Reduced disease progression and increased survival in treated hamsters compared to control hamsters

(156)


example, a study published in the Journal of Neuropathology and Experimental Neurology found that the transplantation of neural stem cells into the brains of CJD-afflicted mice led to significant improvements in their motor functions and cognitive abilities. The transplanted cells not only survived in the brain, but also differentiated into various types of neural cells and integrated with the existing brain tissue (152).

One of the challenges in using neural stem cell therapy for CJD is the risk of transmission of the disease. CJD is caused by prions, abnormal proteins that can trigger misfolding of

normal proteins in the brain (8, 153). If the donor neural stem cells are contaminated with these prions, they could potentially spread the disease to the recipient. Therefore, extensive screening and safety measures need to be in place to minimize this risk [150].

Despite these challenges, the potential benefits of neural stem cell therapy for CJD are too significant to ignore. More research is needed to fully understand the safety and efficacy of this therapy, but the initial results are very encouraging. If successful, neural stem cell therapy could offer an effective and long-lasting treatment for this devastating disease, and even possibly pave the way for similar therapies for other neurodegenerative disorders.

These studies in Table 9 were conducted in animal models of prion diseases, which include CJD, but their relevance to human CJD is not yet clear. Further research is needed to determine whether neural stem cell therapy could be effective in humans with CJD.

 

Conclusion

Neurodegenerative diseases are a group of debilitating conditions that currently have no known cure. However, the use of Neural Stem Cell Therapy has emerged as a promising treatment option for these conditions. Neural Stem Cells possess the unique ability to regenerate lost or damaged neurons in the brain, which could potentially slow or reverse the effects of neurodegenerative diseases. This review highlights the current research and clinical trials investigating Neural Stem Cell Therapy's efficacy and explores its benefits and limitations.

Several studies have shown that Neural Stem Cell Therapy may have benefits for patients with neurodegenerative disorders. For example, a clinical trial involving patients with

Parkinson's disease found that Neural Stem Cell Therapy significantly improved motor function and was well-tolerated by patients. Similarly, a study examining the use of Neural Stem Cells in Alzheimer's disease demonstrated reduced amyloid-beta plaque and improved cognitive function in patients.

 

Although promising, there are still limitations to this therapeutic approach. One significant challenge is optimizing the delivery of Neural Stem Cells to the appropriate area of the brain or spinal cord affected by the disease. Additionally, there are concerns around the safety and potential for graft rejection.

Overall, while it is still early in the development of Neural Stem Cell Therapy for neurodegenerative diseases, the results from these studies and clinical trials are promising. Neural Stem Cells have the unique ability to regenerate damaged or lost neurons in the brain, which could slow or reverse the effects of neurodegenerative diseases. Further research is needed to establish the safety and efficacy of Neural Stem Cell Therapy in treating these debilitating conditions. Despite these limitations, the use of Neural Stem Cells provides a new avenue to explore and could potentially lead to the development of more effective treatments for neurodegenerative diseases.

 

Discussion

Neural Stem Cell Therapy is a rapidly advancing field in medicine that shows immense potential in the treatment of various neurodegenerative diseases. Recent studies have demonstrated promising results in the use of Neural Stem Cell Therapy for Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, ALS, spinocerebellar ataxia, frontotemporal dementia, Creutzfeldt-Jakob disease, and Lewy body disease.

Compared to other types of stem cells, such as mesenchymal and hematopoietic stem cells, neural stem cells have the unique ability to differentiate into different types of neural cells in the brain and spinal cord. Furthermore, they have been found to reduce inflammation, promote the formation of new neurons, and facilitate functional recovery in degenerating neurological systems.

While further research is needed to optimize neural stem cell therapy for each of these diseases, the progress made so far is significant. These therapies offer new hope for patients with various neurodegenerative disorders and represent an exciting breakthrough in the field of medicine.

In conclusion, Neural Stem Cell Therapy holds significant potential for treating various neurodegenerative diseases. Ongoing studies and clinical trials continue to provide promising results, offering hope and new avenues for treatment for millions of people around the world who suffer from these debilitating diseases. This is a cause for celebration in the field of medicine.

 

Abbreviations

Alzheimer's disease: AD

Parkinson's disease: PD

Huntington's disease: HD

Multiple sclerosis: MS

Amyotrophic Lateral Sclerosis: ALS

Spinocerebellar ataxia: SCA

Frontotemporal dementia: FTD

Creutzfeldt-Jakob disease: CJD

Lewy body disease: LBD

NSC: Neural Stem Cell

 

Deceleration

 

Ethics approval and consent to participate

Not applicable

Consent for publication

Not applicable

Competing interests

Not applicable

Funding

Not applicable

Author’s contributions

Idea: Dr. Mehrdad Gholamzad, Amir Gholamzad

Introduction: Dr. Mehrdad Gholamzad, Amir Gholamzad, Omid Moeini, Seyed Armit Hosseini

Alzheimer's disease: Dr. Mehrdad Gholamzad, Amir Gholamzad, Hadis Sadeghi, Maryam Azizabadi farahani

Parkinson's disease: Dr. Mehrdad Gholamzad, Amir Gholamzad, Hadis Sadeghi, Ali Faraji

Huntington's disease: Dr. Mehrdad Gholamzad, Amir Gholamzad, Hadis Sdeghi, Sajad Khonche

multiple sclerosis: Dr. Mehrdad Gholamzad, Amir Gholamzad, Hadis Sadeghi, Mahya Aarasterad Amyotrophic Lateral Sclerosis: Dr. Mehrdad Gholamzad, Amir Gholamzad, Hadis Sadeghi, Mahsa Khatibi

spinocerebellar ataxia: Dr. Mehrdad Gholamzad, Amir Gholamzad, Mohammadmatin Noruikhani

frontotemporal dementia: Dr. Mehrdad Gholamzad, Amir Gholamzad, Hadis Sadeghi

Creutzfeldt-Jakob disease: Dr. Mehrdad Gholamzad, Amir Gholamzad, Hadis Sadeghi

Lewy body disease: Dr. Mehrdad Gholamzad, Amir Gholamzad, Hadis Sadeghi

Conclusion: Dr. Mehrdad Gholamzad

Acknowledgements

Not applicable

Availability of data and materials

Not applicable

Consent for publication

This manuscript has been approved for publication by all authors.

 
1. Teleanu DM, Niculescu A-G, Lungu II, Radu CI, Vladâcenco O, Roza E, et al. An overview of oxidative stress, neuroinflammation, and neurodegenerative diseases. 2022;23(11):5938.
https://doi.org/10.3390/ijms23115938
PMid:35682615 PMCid:PMC9180653
 
2. Esmaeili Y, Yarjanli Z, Pakniya F, Bidram E, Łos MJ, Eshraghi M, et al. Targeting autophagy, oxidative stress, and ER stress for neurodegenerative disease treatment. 2022;345:147-75.
https://doi.org/10.1016/j.jconrel.2022.03.001
PMid:35248646
 
3. Zhou J, Ni W, Ling Y, Lv X, Niu D, Zeng Y, et al. Human neural stem cell secretome inhibits lipopolysaccharide-induced neuroinflammation through modulating microglia polarization by activating peroxisome proliferator-activated receptor gamma. 2022;31(13-14):369-82.
https://doi.org/10.1089/scd.2022.0081
PMid:35481777
 
4. Kopach OJWJoSC. Monitoring maturation of neural stem cell grafts within a host microenvironment. 2019;11(11):982.
https://doi.org/10.4252/wjsc.v11.i11.982
PMid:31768224 PMCid:PMC6851006
 
5. Zhao L, Liu J-W, Shi H-Y, Ma Y-MJWJoSC. Neural stem cell therapy for brain disease. 2021;13(9):1278.
https://doi.org/10.4252/wjsc.v13.i9.1278
PMid:34630862 PMCid:PMC8474718
 
6. Gilbert EAB, Lakshman N, Lau KSK, Morshead CM. Regulating Endogenous Neural Stem Cell Activation to Promote Spinal Cord Injury Repair. Cells. 2022;11(5).
https://doi.org/10.3390/cells11050846
PMid:35269466 PMCid:PMC8909806
 
7. Nicaise AM, D'Angelo A, Ionescu R-B, Krzak G, Willis CM, Pluchino SJC, et al. The role of neural stem cells in regulating glial scar formation and repair. 2022:1-16.
 
8. De Gioia R, Biella F, Citterio G, Rizzo F, Abati E, Nizzardo M, et al. Neural stem cell transplantation for neurodegenerative diseases. 2020;21(9):3103.
https://doi.org/10.3390/ijms21093103
PMid:32354178 PMCid:PMC7247151
 
9. Walton C, King R, Rechtman L, Kaye W, Leray E, Marrie RA, et al. Rising prevalence of multiple sclerosis worldwide: Insights from the Atlas of MS. 2020;26(14):1816-21.
https://doi.org/10.1177/1352458520970841
PMid:33174475 PMCid:PMC7720355
 
10. Rahmanzadeh R, Lu P-J, Barakovic M, Weigel M, Maggi P, Nguyen TD, et al. Myelin and axon pathology in multiple sclerosis assessed by myelin water and multi-shell diffusion imaging. 2021;144(6):1684-96.
https://doi.org/10.1093/brain/awab088
PMid:33693571 PMCid:PMC8374972
 
11. Giovannoni G, Mathews JJN, therapy. Cladribine Tablets for Relapsing-Remitting Multiple Sclerosis: A Clinician's Review. 2022;11(2):571-95.
https://doi.org/10.1007/s40120-022-00339-7
PMid:35318617 PMCid:PMC8940595
 
12. Pluchino S, Smith JA, Peruzzotti-Jametti LJTiMM. Promises and limitations of neural stem cell therapies for progressive multiple sclerosis. 2020;26(10):898-912.
https://doi.org/10.1016/j.molmed.2020.04.005
PMid:32448751
 
13. Gordon J, Amini SJNCCM, Protocols. General overview of neuronal cell culture. 2021:1-8.
https://doi.org/10.1007/978-1-0716-1437-2_1
PMid:34033073
 
14. Willis CM, Nicaise AM, Peruzzotti-Jametti L, Pluchino SJBr. The neural stem cell secretome and its role in brain repair. 2020;1729:146615.
https://doi.org/10.1016/j.brainres.2019.146615
PMid:31863730
 
15. Puranik N, Arukha AP, Yadav SK, Yadav D, Jin JOJCSCR, Therapy. Exploring the role of stem cell therapy in treating neurodegenerative diseases: Challenges and current perspectives. 2022;17(2):113-25.
https://doi.org/10.2174/1574888X16666210810103838
PMid:35135462
 
16. Willis CM, Nicaise AM, Hamel R, Pappa V, Peruzzotti-Jametti L, Pluchino SJFicn. Harnessing the neural stem cell secretome for regenerative neuroimmunology. 2020;14:590960.
https://doi.org/10.3389/fncel.2020.590960
PMid:33250716 PMCid:PMC7674923
 
17. Rahman MM, Islam MR, Islam MT, Harun-Or-Rashid M, Islam M, Abdullah S, et al. Stem cell transplantation therapy and neurological disorders: current status and future perspectives. 2022;11(1):147.
https://doi.org/10.3390/biology11010147
PMid:35053145 PMCid:PMC8772847
 
18. Bezukladova S, Genchi A, Panina-Bordignon P, Martino GJCOiN. Promoting exogenous repair in multiple sclerosis: myelin regeneration. 2022;35(3):313-8.
https://doi.org/10.1097/WCO.0000000000001062
PMid:35674074
 
19. Smith C, Gore A, Yan W, Abalde-Atristain L, Li Z, He C, et al. Whole-genome sequencing analysis reveals high specificity of CRISPR/Cas9 and TALEN-based genome editing in human iPSCs. 2014;15(1):12-3.
https://doi.org/10.1016/j.stem.2014.06.011
PMid:24996165 PMCid:PMC4338993
 
20. Chhabra JS, Balaji SS, Singh A, Mishra S, Ganpule AP, Sabnis RB, et al. Urethral balloon dilatation: factors affecting outcomes. 2016;96(4):427-31.
https://doi.org/10.1159/000443704
PMid:26845345
 
21. Mancardi G, Saccardi RJTLN. Autologous haematopoietic stem-cell transplantation in multiple sclerosis. 2008;7(7):626-36.
https://doi.org/10.1016/S1474-4422(08)70138-8
PMid:18565456
 
22. Connick P, Kolappan M, Crawley C, Webber DJ, Patani R, Michell AW, et al. Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open-label phase 2a proof-of-concept study. 2012;11(2):150-6.
https://doi.org/10.1016/S1474-4422(11)70305-2
PMid:22236384
 
23. Jhanji R, Behl T, Sehgal A, Bungau SJM. Mitochondrial dysfunction and traffic jams in amyotrophic lateral sclerosis. 2021;58:102-10.
https://doi.org/10.1016/j.mito.2021.02.008
PMid:33639271
 
24. Ustyantseva E, Medvedev S, Zakian SJMoGP, Repair. Studying ALS: Current approaches, effect on potential treatment strategy. 2020:195-217.
https://doi.org/10.1007/978-3-030-41283-8_11
PMid:32383122
 
25. Ahani‐Nahayati M, Shariati A, Mahmoodi M, Olegovna Zekiy A, Javidi K, Shamlou S, et al. Stem cell in neurodegenerative disorders; an emerging strategy. 2021;81(4):291-311.
https://doi.org/10.1002/jdn.10101
PMid:33650716
 
26. Lin B, Lu L, Wang Y, Zhang Q, Wang Z, Cheng G, et al. Nanomedicine directs neuronal differentiation of neural stem cells via silencing long noncoding RNA for stroke therapy. 2021;21(1):806-15.
https://doi.org/10.1021/acs.nanolett.0c04560
PMid:33395306
 
27. Francesca S, De Marchi F, Mazzini L, Bendotti CJBRB. CELL THERAPY IN ALS: an update on preclinical and clinical studies. 2023.
 
28. Zhu Q, Lu PJSC-bTfND. Stem cell transplantation for amyotrophic lateral sclerosis. 2020:71-97.
https://doi.org/10.1007/978-981-15-4370-8_6
PMid:33105496
 
29. Je G, Keyhanian K, Ghasemi MJNR. Overview of stem cells therapy in amyotrophic lateral sclerosis. 2021;43(8):616-32.
https://doi.org/10.1080/01616412.2021.1893564
PMid:33632084
 
30. Morata-Tarifa C, Azkona G, Glass J, Mazzini L, Sanchez-Pernaute RJNRM. Looking backward to move forward: a meta-analysis of stem cell therapy in amyotrophic lateral sclerosis. 2021;6(1):20.
https://doi.org/10.1038/s41536-021-00131-5
PMid:33795700 PMCid:PMC8016966
 
31. Ediriweera GR, Chen L, Yerbury JJ, Thurecht KJ, Vine KLJMP. Non-viral vector-mediated gene therapy for ALS: Challenges and future perspectives. 2021;18(6):2142-60.
https://doi.org/10.1021/acs.molpharmaceut.1c00297
PMid:34010004
 
32. Dhasmana S, Dhasmana A, Narula AS, Jaggi M, Yallapu MM, Chauhan SCJLS. The panoramic view of amyotrophic lateral sclerosis: A fatal intricate neurological disorder. 2022;288:120156.
https://doi.org/10.1016/j.lfs.2021.120156
PMid:34801512
 
33. Bonaventura G, Munafò A, Bellanca CM, La Cognata V, Iemmolo R, Attaguile GA, et al. Stem cells: innovative therapeutic options for neurodegenerative diseases? 2021;10(8):1992.
https://doi.org/10.3390/cells10081992
PMid:34440761 PMCid:PMC8391848
 
34. Boulis NM, Federici T, Glass JD, Lunn JS, Sakowski SA, Feldman ELJNRN. Translational stem cell therapy for amyotrophic lateral sclerosis. 2012;8(3):172-6.
https://doi.org/10.1038/nrneurol.2011.191
PMid:22158518
 
35. Liu Z, Jiang Y, Li X, Hu ZJSCR. Embryonic stem cell-derived peripheral auditory neurons form neural connections with mouse central auditory neurons in vitro via the α2δ1 receptor. 2018;11(1):157-70.
https://doi.org/10.1016/j.stemcr.2018.05.006
PMid:29887365 PMCid:PMC6066995
 
36. Ciervo Y, Ning K, Jun X, Shaw PJ, Mead RJJMn. Advances, challenges and future directions for stem cell therapy in amyotrophic lateral sclerosis. 2017;12(1):1-22.
https://doi.org/10.1186/s13024-017-0227-3
PMid:29132389 PMCid:PMC5683324
 
37. Xu M, Nie Y, Yang Y, Lu Y-T, Su QJTn. Risk of neurological toxicities following the use of different immune checkpoint inhibitor regimens in solid tumors: a systematic review and meta-analysis. 2019;24(3):75-83.
https://doi.org/10.1097/NRL.0000000000000230
PMid:31045716
 
38. Castelli V, d'Angelo M, Quintiliani M, Benedetti E, Cifone MG, Cimini AJNRR. The emerging role of probiotics in neurodegenerative diseases: new hope for Parkinson's disease? 2021;16(4):628.
https://doi.org/10.4103/1673-5374.295270
PMid:33063712 PMCid:PMC8067943
 
39. Berlet R, Galang Cabantan DA, Gonzales-Portillo D, Borlongan CVJFiC, Biology D. Enriched environment and exercise enhance stem cell therapy for stroke, Parkinson's disease, and Huntington's disease. 2022;10:322.
https://doi.org/10.3389/fcell.2022.798826
PMid:35309929 PMCid:PMC8927702
 
40. Cha Y, Park T-Y, Leblanc P, Kim K-SJJoMD. Current Status and Future Perspectives on Stem Cell-Based Therapies for Parkinson's Disease. 2023.
https://doi.org/10.14802/jmd.22141
PMid:36628428 PMCid:PMC9978267
 
41. SHAH AA, KASHYAP K, GUPTA AJIJoPR. Parkinson's Disease: An Introduction to the Causes and Management Strategies. 2020;12(4).
https://doi.org/10.31838/ijpr/2020.12.04.698
 
42. Fischer I, Dulin JN, Lane MAJNRN. Transplanting neural progenitor cells to restore connectivity after spinal cord injury. 2020;21(7):366-83.
https://doi.org/10.1038/s41583-020-0314-2
PMid:32518349 PMCid:PMC8384139
 
43. Al-Mayyahi RJBoPSA. NEURAL STEM CELLS AS A PROMISING THERAPEUTIC APPROACH IN NEUROLOGICAL DISEASES. 2021;44(2):525-36.
https://doi.org/10.21608/bfsa.2021.207179
 
44. Zeng X, Qin HJA, Disease. Stem Cell Transplantation for Parkinson's Disease: Current Challenges and Perspectives. 2022;13(6).
https://doi.org/10.14336/AD.2022.0312
PMid:36465172 PMCid:PMC9662280
 
45. Kim SW, Woo H-J, Kim EH, Kim HS, Suh HN, Kim S-h, et al. Neural stem cells derived from human midbrain organoids as a stable source for treating Parkinson's disease: Midbrain organoid-NSCs (Og-NSC) as a stable source for PD treatment. 2021;204:102086.
https://doi.org/10.1016/j.pneurobio.2021.102086
PMid:34052305
 
46. Hayashi Y, Lin H-T, Lee C-C, Tsai K-JJJobs. Effects of neural stem cell transplantation in Alzheimer's disease models. 2020;27:1-11.
https://doi.org/10.1186/s12929-020-0622-x
PMid:31987051 PMCid:PMC6986162
 
47. Pereira MC, Boese AC, Murad R, Yin J, Hamblin MH, Lee J-PJEN. Reduced dopaminergic neuron degeneration and global transcriptional changes in Parkinson's disease mouse brains engrafted with human neural stems during the early disease stage. 2022;352:114042.
https://doi.org/10.1016/j.expneurol.2022.114042
PMid:35271839
 
48. Aarsland D, Batzu L, Halliday GM, Geurtsen GJ, Ballard C, Ray Chaudhuri K, et al. Parkinson disease-associated cognitive impairment. 2021;7(1):47.
https://doi.org/10.1038/s41572-021-00280-3
PMid:34210995
 
49. Kim TW, Koo SY, Studer LJFic, biology d. Pluripotent stem cell therapies for Parkinson disease: present challenges and future opportunities. 2020;8:729.
https://doi.org/10.3389/fcell.2020.00729
PMid:32903681 PMCid:PMC7438741
 
50. Xiao Z, Lei T, Liu Y, Yang Y, Bi W, Du HJSCR, et al. The potential therapy with dental tissue-derived mesenchymal stem cells in Parkinson's disease. 2021;12(1):1-11.
https://doi.org/10.1186/s13287-020-01957-4
PMid:33407864 PMCid:PMC7789713
 
51. Jang SE, Qiu L, Chan LL, Tan E-K, Zeng LJFin. Current status of stem cell-derived therapies for Parkinson's disease: from cell assessment and imaging modalities to clinical trials. 2020;14:558532.
https://doi.org/10.3389/fnins.2020.558532
PMid:33177975 PMCid:PMC7596695
 
52. Yefroyev DA, Jin SJB. Induced Pluripotent Stem Cells for Treatment of Alzheimer's and Parkinson's Diseases. 2022;10(2):208.
https://doi.org/10.3390/biomedicines10020208
PMid:35203418 PMCid:PMC8869146
 
53. Yamanaka SJCsc. Pluripotent stem cell-based cell therapy-promise and challenges. 2020;27(4):523-31.
https://doi.org/10.1016/j.stem.2020.09.014
PMid:33007237
 
54. Christine CW, Richardson RM, Van Laar AD, Thompson ME, Fine EM, Khwaja OS, et al. Safety of AADC gene therapy for moderately advanced Parkinson disease: three-year outcomes from the PD-1101 trial. 2022;98(1):e40-e50.
https://doi.org/10.1212/WNL.0000000000012952
PMid:34649873 PMCid:PMC8726573
 
55. Tomishima M, Kirkeby AJJoPsD. Bringing advanced therapies for Parkinson's disease to the clinic: The Scientist's Perspective. 2021;11(s2):S135-S40.
https://doi.org/10.3233/JPD-212685
PMid:34250954 PMCid:PMC8543246
 
56. Tolosa E, Wenning G, Poewe WJTLN. The diagnosis of Parkinson's disease. 2006;5(1):75-86.
https://doi.org/10.1016/S1474-4422(05)70285-4
PMid:16361025
 
57. Maiese KJCNR. Biomarkers for Parkinson's Disease and Neurodegenerative Disorders: A Role for Non-coding RNAs. 2022;19(2):127-30.
https://doi.org/10.2174/1567202619666220602125806
PMid:35657043
 
58. Schneider SA, Alcalay RNJJon. Precision medicine in Parkinson's disease: emerging treatments for genetic Parkinson's disease. 2020;267:860-9.
https://doi.org/10.1007/s00415-020-09705-7
PMid:31974807 PMCid:PMC7035220
 
59. Król-Grzymała A, Sienkiewicz-Szłapka E, Fiedorowicz E, Rozmus D, Cieślińska A, Grzybowski AJIJoMS. Tear Biomarkers in Alzheimer's and Parkinson's Diseases, and Multiple Sclerosis: Implications for Diagnosis (Systematic Review). 2022;23(17):10123.
https://doi.org/10.3390/ijms231710123
PMid:36077520 PMCid:PMC9456033
 
60. Ritter VC, Bonsaksen TJJoMH. Improvement in quality of life following a multidisciplinary rehabilitation program for patients with Parkinson's disease. 2019:219-27.
https://doi.org/10.2147/JMDH.S202827
PMid:31114215 PMCid:PMC6497500
 
61. Carroll LM, Morris M, O'Connor WT, Volpe D, Salsberg J, Clifford AMJJoPsD. Evidence-Based Aquatic Therapy Guidelines for Parkinson's Disease: An International Consensus Study. 2022;12(2):621-37.
https://doi.org/10.3233/JPD-212881
PMid:34842200
 
62. Garitaonandia I, Gonzalez R, Christiansen-Weber T, Abramihina T, Poustovoitov M, Noskov A, et al. Neural stem cell tumorigenicity and biodistribution assessment for phase I clinical trial in Parkinson's disease. 2016;6(1):1-12.
https://doi.org/10.1038/srep34478
PMid:27686862 PMCid:PMC5055076
 
63. Korczyn AD, Reichmann H, Tolosa EJJotns. Idiopathic Parkinson's syndrome. Preface. 2011. p. 1.
https://doi.org/10.1016/j.jns.2011.08.011
PMid:21893323
 
64. Tian S-m, Ma Y-x, Shi J, Lou T-y, Liu S-s, Li G-yJNRR. Acrylamide neurotoxicity on the cerebrum of weaning rats. 2015;10(6):938.
https://doi.org/10.4103/1673-5374.158357
PMid:26199611 PMCid:PMC4498356
 
65. Madrazo I, Kopyov O, Ávila-Rodríguez M, Ostrosky F, Carrasco H, Kopyov A, et al. Transplantation of human neural progenitor cells (NPC) into putamina of parkinsonian patients: a case series study, safety and efficacy four years after surgery. 2019;28(3):269-85.
https://doi.org/10.1177/0963689718820271
PMid:30574805 PMCid:PMC6425108
 
66. Apodaca LA, Baddour AAD, Garcia C, Alikhani L, Giedzinski E, Ru N, et al. Human neural stem cell-derived extracellular vesicles mitigate hallmarks of Alzheimer's disease. 2021;13(1):1-18.
https://doi.org/10.1186/s13195-021-00791-x
PMid:33676561 PMCid:PMC7937214
 
67. John OO, Amarachi IS, Chinazom AP, Adaeze E, Kale MB, Umare MD, et al. Phytotherapy: A promising approach for the treatment of Alzheimer's disease. 2022;2:100030.
https://doi.org/10.1016/j.prmcm.2021.100030
 
68. Yiannopoulou KG, Papageorgiou SGJJocnsd. Current and future treatments in Alzheimer disease: an update. 2020;12:1179573520907397.
https://doi.org/10.1177/1179573520907397
PMid:32165850 PMCid:PMC7050025
 
69. Srivastava S, Ahmad R, Khare SKJEJoMC. Alzheimer's disease and its treatment by different approaches: A review. 2021;216:113320.
https://doi.org/10.1016/j.ejmech.2021.113320
PMid:33652356
 
70. Liu Q, Zhang L, Zhang JJB, Pharmacotherapy. Induced pluripotent stem cell-derived neural progenitor cell transplantation promotes regeneration and functional recovery after post-traumatic stress disorder in rats. 2021;133:110981.
https://doi.org/10.1016/j.biopha.2020.110981
PMid:33186796
 
71. Wang W, Zhao F, Ma X, Perry G, Zhu XJMN. Mitochondria dysfunction in the pathogenesis of Alzheimer's disease: Recent advances. 2020;15:1-22.
https://doi.org/10.1186/s13024-020-00376-6
PMid:32471464 PMCid:PMC7257174
 
72. Drexler HJScfttondN. Monitoring of bone marrow cell homing into 37. Lindvall, O.; Kokaia, Z. 2006;441:1094-6.
https://doi.org/10.1038/nature04960
PMid:16810245
 
73. Mihardja M, Roy J, Wong KY, Aquili L, Heng BC, Chan YS, et al. Therapeutic potential of neurogenesis and melatonin regulation in Alzheimer's disease. 2020;1478(1):43-62.
https://doi.org/10.1111/nyas.14436
PMid:32700392
 
74. Hinkle J. Cranial Irradiation-mediated Dendritic Spine Loss Is Microglial Complement Receptor 3-Dependent, Sex-specific, and Associated with Cognitive Impairment: University of Rochester; 2020.
 
75. DAVISON LR. Investigating the Effects of FTY720 on Spatial Memory and Neuroinflammation in a Rodent Model of Alzheimer's Disease: Trinity College Dublin. School of Medicine. Discipline of Physiology; 2020.
 
76. Kim TA, Syty MD, Wu K, Ge SJZR. Adult hippocampal neurogenesis and its impairment in Alzheimer's disease. 2022;43(3):481.
https://doi.org/10.24272/j.issn.2095-8137.2021.479
PMid:35503338 PMCid:PMC9113964
 
77. Pous L, Deshpande SS, Nath S, Mezey S, Malik SC, Schildge S, et al. Fibrinogen induces neural stem cell differentiation into astrocytes in the subventricular zone via BMP signaling. 2020;11(1):630.
https://doi.org/10.1038/s41467-020-14466-y
PMid:32005867 PMCid:PMC6994610
 
78. Sung P-S, Lin P-Y, Liu C-H, Su H-C, Tsai K-JJIjoms. Neuroinflammation and neurogenesis in Alzheimer's disease and potential therapeutic approaches. 2020;21(3):701.
https://doi.org/10.3390/ijms21030701
PMid:31973106 PMCid:PMC7037892
 
79. Kim GS, Hwang KA, Choi KC. A promising therapeutic strategy for metastatic gestational trophoblastic disease: Engineered anticancer gene-expressing stem cells to selectively target choriocarcinoma. Oncology letters. 2019;17(3):2576-82.
https://doi.org/10.3892/ol.2019.9911
PMid:30867726 PMCid:PMC6396211
 
80. Kang JM, Yeon BK, Cho S-J, Suh Y-HJJoAsD. Stem cell therapy for Alzheimer's disease: a review of recent clinical trials. 2016;54(3):879-89.
https://doi.org/10.3233/JAD-160406
PMid:27567851
 
81. McGinley LM, Kashlan ON, Bruno ES, Chen KS, Hayes JM, Kashlan SR, et al. Human neural stem cell transplantation improves cognition in a murine model of Alzheimer's disease. 2018;8(1):14776.
https://doi.org/10.1038/s41598-018-33017-6
PMid:30283042 PMCid:PMC6170460
 
82. Sah SK, Kim HY, Lee JH, Lee S-W, Kim H-S, Kim Y-S, et al. Effects of human mesenchymal stem cells coculture on calcium-induced differentiation of normal human keratinocytes. 2017;35(6):1592-602.
https://doi.org/10.1002/stem.2593
PMid:28207189
 
83. Lai X, Wen H, Li Y, Lu L, Tang CJFiAN. The comparative efficacy of multiple interventions for mild cognitive impairment in Alzheimer's Disease: a bayesian network meta-analysis. 2020;12:121.
https://doi.org/10.3389/fnagi.2020.00121
PMid:32581760 PMCid:PMC7289916
 
84. Zhang F-Q, Jiang J-L, Zhang J-T, Niu H, Fu X-Q, Zeng L-LJ中. 干细胞治疗阿尔茨海默病的现状及前景. 2020;15(2):242.
 
85. Lu M-H, Ji W-L, Chen H, Sun Y-Y, Zhao X-Y, Wang F, et al. Intranasal transplantation of human neural stem cells ameliorates Alzheimer's disease-like pathology in a mouse model. 2021;13:650103.
https://doi.org/10.3389/fnagi.2021.650103
PMid:33776747 PMCid:PMC7987677
 
86. Monk R, Connor BJSC-bTfND. Cell Replacement Therapy for Huntington's Disease. 2020:57-69.
https://doi.org/10.1007/978-981-15-4370-8_5
PMid:33105495
 
87. Rocha NP, Charron O, Latham LB, Colpo GD, Zanotti-Fregonara P, Yu M, et al. Microglia activation in basal ganglia is a late event in Huntington disease pathophysiology. 2021;8(3).
https://doi.org/10.1212/NXI.0000000000000984
PMid:33795375 PMCid:PMC8017723
 
88. Irfan Z, Khanam S, Karmakar V, Firdous SM, El Khier BSIA, Khan I, et al. Pathogenesis of Huntington's Disease: An Emphasis on Molecular Pathways and Prevention by Natural Remedies. 2022;12(10):1389.
https://doi.org/10.3390/brainsci12101389
PMid:36291322 PMCid:PMC9599635
 
89. Park HJ, Han A, Kim JY, Choi J, Bae HS, Cho G-b, et al. SUPT4H1-edited stem cell therapy rescues neuronal dysfunction in a mouse model for Huntington's disease. 2022;7(1):8.
https://doi.org/10.1038/s41536-022-00219-6
PMid:35449161 PMCid:PMC9023523
 
90. Ji X, Zhou Y, Gao Q, He H, Wu Z, Feng B, et al. Functional reconstruction of the basal ganglia neural circuit by human striatal neurons in hypoxic-ischaemic injured brain. 2023;146(2):612-28.
https://doi.org/10.1093/brain/awac358
PMid:36516880 PMCid:PMC9924911
 
91. Besusso D, Schellino R, Boido M, Belloli S, Parolisi R, Conforti P, et al. Stem cell-derived human striatal progenitors innervate striatal targets and alleviate sensorimotor deficit in a rat model of Huntington disease. 2020;14(5):876-91.
https://doi.org/10.1016/j.stemcr.2020.03.018
PMid:32302555 PMCid:PMC7220987
 
92. Colpo GD, Stimming EF, Teixeira ALJM, Neuroscience C. Stem cells in animal models of Huntington disease: A systematic review. 2019;95:43-50.
https://doi.org/10.1016/j.mcn.2019.01.006
PMid:30685323
 
93. disease SHCJJoHs. Stem Cells for Huntington's disease (SC4HD): An international consortium to facilitate stem cell-based therapy for Huntington's disease. 2021;10(2):221-6.
https://doi.org/10.3233/JHD-210473
PMid:33814456 PMCid:PMC8293637
 
94. Lange J, Wood-Kaczmar A, Ali A, Farag S, Ghosh R, Parker J, et al. Mislocalization of nucleocytoplasmic transport proteins in human Huntington's disease PSC-derived striatal neurons. 2021;15:742763.
https://doi.org/10.3389/fncel.2021.742763
PMid:34658796 PMCid:PMC8519404
 
95. Yoon Y, Kim HS, Jeon I, Noh J-E, Park HJ, Lee S, et al. Implantation of the clinical-grade human neural stem cell line, CTX0E03, rescues the behavioral and pathological deficits in the quinolinic acid-lesioned rodent model of Huntington's disease. 2020;38(8):936-47.
https://doi.org/10.1002/stem.3191
PMid:32374064 PMCid:PMC7496241
 
96. Kageyama R, Shimojo H, Ohtsuka TJNr. Dynamic control of neural stem cells by bHLH factors. 2019;138:12-8.
https://doi.org/10.1016/j.neures.2018.09.005
PMid:30227160
 
97. Cho IK, Hunter CE, Ye S, Pongos AL, Chan AWSJNRm. Combination of stem cell and gene therapy ameliorates symptoms in Huntington's disease mice. 2019;4(1):7.
https://doi.org/10.1038/s41536-019-0066-7
PMid:30937182 PMCid:PMC6435637
 
98. Jareño PG, Bartley OJ, Precious SV, Rosser AE, Lelos MJJIRoN. Challenges in progressing cell therapies to the clinic for Huntington's disease: A review of the progress made with pluripotent stem cell derived medium spiny neurons. 2022;166:1-48.
https://doi.org/10.1016/bs.irn.2022.09.003
PMid:36424090
 
99. Bartley OJ, Lelos MJ, Gray WP, Rosser AEJNS. Do foetal transplant studies continue to be justified in Huntington's disease? 2021;5(4):NS20210019.
https://doi.org/10.1042/NS20210019
PMid:34956650 PMCid:PMC8674623
 
100. Rosser AE, Busse ME, Gray WP, Badin RA, Perrier AL, Wheelock V, et al. Translating cell therapies for neurodegenerative diseases: Huntington's disease as a model disorder. 2022;145(5):1584-97.
https://doi.org/10.1093/brain/awac086
PMid:35262656 PMCid:PMC9166564
 
101. Blazek P, Davis SL, Greenberg BM, Conger A, Conger D, Vernino S, et al. Objective characterization of the relative afferent pupillary defect in MS. 2012;323(1-2):193-200.
https://doi.org/10.1016/j.jns.2012.09.015
PMid:23026533
 
102. Rega D, Pecori B, Scala D, Avallone A, Pace U, Petrillo A, et al. Evaluation of tumor response after short-course radiotherapy and delayed surgery for rectal cancer. 2016;11(8):e0160732.
https://doi.org/10.1371/journal.pone.0160732
PMid:27548058 PMCid:PMC4993446
 
103. Sadan O, Shemesh N, Barzilay R, Dadon-Nahum M, Blumenfeld-Katzir T, Assaf Y, et al. Mesenchymal stem cells induced to secrete neurotrophic factors attenuate quinolinic acid toxicity: a potential therapy for Huntington's disease. 2012;234(2):417-27.
https://doi.org/10.1016/j.expneurol.2011.12.045
PMid:22285250
 
104. Klockgether T, Mariotti C, Paulson HLJNrDp. Spinocerebellar ataxia. 2019;5(1):24.
https://doi.org/10.1038/s41572-019-0074-3
PMid:30975995
 
105. Triangto K, Setiono S, Purba HB. International Classification of Functioning, Health and Disability (ICF) Conceptual Approach towards Spinocerebellar Ataxia: IntechOpen; 2021.
https://doi.org/10.5772/intechopen.96275
 
106. He L, Chen Z, Peng L, Tang B, Jiang HJEN. Human stem cell models of polyglutamine diseases: sources for disease models and cell therapy. 2021;337:113573.
https://doi.org/10.1016/j.expneurol.2020.113573
PMid:33347831
 
107. Mitoma H, Buffo A, Gelfo F, Guell X, Fucà E, Kakei S, et al. Consensus paper. Cerebellar reserve: from cerebellar physiology to cerebellar disorders. 2020;19:131-53.
https://doi.org/10.1007/s12311-019-01091-9
PMid:31879843 PMCid:PMC6978437
 
108. Nam Y, Yoon D, Hong J, Kim MS, Lee TY, Kim KS, et al. Therapeutic effects of human mesenchymal stem cells in a mouse model of cerebellar ataxia with neuroinflammation. 2020;9(11):3654.
https://doi.org/10.3390/jcm9113654
PMid:33202913 PMCid:PMC7698164
 
109. Park N, Sharma C, Jung UJ, Kim S, Nam Y, Kim K-S, et al. Mesenchymal Stem Cell Transplantation Ameliorates Ara-C-Induced Motor Deficits in a Mouse Model of Cerebellar Ataxia. 2023;12(5):1756.
https://doi.org/10.3390/jcm12051756
PMid:36902541 PMCid:PMC10003478
 
110. Negredo PN, Yeo RW, Brunet AJCsc. Aging and rejuvenation of neural stem cells and their niches. 2020;27(2):202-23.
https://doi.org/10.1016/j.stem.2020.07.002
PMid:32726579 PMCid:PMC7415725
 
111. Correia JS, Duarte-Silva S, Salgado AJ, Maciel PJNRR. Cell-based therapeutic strategies for treatment of spinocerebellar ataxias: an update. 2023;18(6):1203-12.
https://doi.org/10.4103/1673-5374.355981
PMid:36453395 PMCid:PMC9838137
 
112. Marsili L, Sharma J, Outeiro TF, Colosimo CJB. Stem Cell Therapies in Movement Disorders: Lessons from Clinical Trials. 2023;11(2):505.
https://doi.org/10.3390/biomedicines11020505
PMid:36831041 PMCid:PMC9953050
 
113. Bhartiya M, Kumar A, Singh RK, Radhakrishnan DM, Rajan R, Srivastava AKJTC. Mesenchymal Stem Cell Therapy in the Treatment of Neurodegenerative Cerebellar Ataxias: a Systematic Review and Meta-analysis. 2022:1-7.
https://doi.org/10.1007/s12311-022-01403-6
PMid:35451803
 
114. Saeedi P, Halabian R, Fooladi AAIJSci. A revealing review of mesenchymal stem cells therapy, clinical perspectives and Modification strategies. 2019;6.
https://doi.org/10.21037/sci.2019.08.11
PMid:31620481 PMCid:PMC6789202
 
115. Liu G, David BT, Trawczynski M, Fessler RGJScr, reports. Advances in pluripotent stem cells: history, mechanisms, technologies, and applications. 2020;16:3-32.
https://doi.org/10.1007/s12015-019-09935-x
PMid:31760627 PMCid:PMC6987053
 
116. Kon T, Tomiyama M, Wakabayashi KJN. Neuropathology of Lewy body disease: Clinicopathological crosstalk between typical and atypical cases. 2020;40(1):30-9.
https://doi.org/10.1111/neup.12597
PMid:31498507
 
117. Tsai Y-A, Liu R-S, Lirng J-F, Yang B-H, Chang C-H, Wang Y-C, et al. Treatment of spinocerebellar ataxia with mesenchymal stem cells: a phase I/IIa clinical study. 2017;26(3):503-12.
https://doi.org/10.3727/096368916X694373
PMid:28195034 PMCid:PMC5657707
 
118. Li L, Li Y, Fan Z, Wang X, Li Z, Wen J, et al. Ascorbic acid facilitates neural regeneration after sciatic nerve crush injury. 2019;13:108.
https://doi.org/10.3389/fncel.2019.00108
PMid:30949031 PMCid:PMC6437112
 
119. Lee JM, Bae J-S, Jin HKJJoVMS. Intracerebellar transplantation of neural stem cells into mice with neurodegeneration improves neuronal networks with functional synaptic transmission. 2010;72(8):999-1009.
https://doi.org/10.1292/jvms.09-0514
PMid:20339259
 
120. Zhang Y, Wu Q, Ren Y, Zhang Y, Feng LJBRB. A53T α-synuclein induces neurogenesis impairment and cognitive dysfunction in line M83 transgenic mice and reduces the proliferation of embryonic neural stem cells. 2022;182:118-29.
https://doi.org/10.1016/j.brainresbull.2022.02.010
PMid:35182691
 
121. Marsh SE, Blurton-Jones MJNi. Neural stem cell therapy for neurodegenerative disorders: the role of neurotrophic support. 2017;106:94-100.
https://doi.org/10.1016/j.neuint.2017.02.006
PMid:28219641 PMCid:PMC5446923
 
122. Goldberg NS. Disrupting Disease Phenotype in Dementia with Lewy Bodies with Neural Stem Cell Transplantation: University of California, Irvine; 2016.
 
123. Rong Y, Liu W, Wang J, Fan J, Luo Y, Li L, et al. Neural stem cell-derived small extracellular vesicles attenuate apoptosis and neuroinflammation after traumatic spinal cord injury by activating autophagy. 2019;10(5):340.
https://doi.org/10.1038/s41419-019-1571-8
PMid:31000697 PMCid:PMC6472377
 
124. Peng Y, Liou B, Lin Y, Mayhew CN, Fleming SM, Sun YJMT-M, et al. iPSC-derived neural precursor cells engineering GBA1 recovers acid β-glucosidase deficiency and diminishes α-synuclein and neuropathology. 2023;29:185-201.
https://doi.org/10.1016/j.omtm.2023.03.007
PMid:37063480 PMCid:PMC10102010
 
125. Park H, Oh J, Shim G, Cho B, Chang Y, Kim S, et al. In vivo neuronal gene editing via CRISPR-Cas9 amphiphilic nanocomplexes alleviates deficits in mouse models of Alzheimer's disease. 2019;22(4):524-8.
https://doi.org/10.1038/s41593-019-0352-0
PMid:30858603
 
126. Jiang S, Wen N, Li Z, Dube U, Del Aguila J, Budde J, et al. Integrative system biology analyses of CRISPR-edited iPSC-derived neurons and human brains reveal deficiencies of presynaptic signaling in FTLD and PSP. 2018;8(1):265.
https://doi.org/10.1038/s41398-018-0319-z
PMid:30546007 PMCid:PMC6293323
 
127. Garcıa-León JA, Cabrera-Socorro A, Eggermont K, Swijsen A, Terryn J, Fazal R, et al. Generation of a human. 2018;1(20):20.
 
128. di Domenico A, Carola G, Calatayud C, Pons-Espinal M, Muñoz JP, Richaud-Patin Y, et al. Patient-specific iPSC-derived astrocytes contribute to non-cell-autonomous neurodegeneration in Parkinson's disease. 2019;12(2):213-29.
https://doi.org/10.1016/j.stemcr.2018.12.011
PMid:30639209 PMCid:PMC6372974
 
129. Barrett MJ, Cloud LJ, Shah H, Holloway KLJERoN. Therapeutic approaches to cholinergic deficiency in Lewy body diseases. 2020;20(1):41-53.
https://doi.org/10.1080/14737175.2020.1676152
PMid:31577469
 
130. Lakatos A, Goldberg NR, Blurton-Jones MJAnc. Integrated analysis of genetic, behavioral, and biochemical data implicates neural stem cell-induced changes in immunity, neurotransmission and mitochondrial function in Dementia with Lewy Body mice. 2017;5(1):1-18.
https://doi.org/10.1186/s40478-017-0421-0
PMid:28283027 PMCid:PMC5345195
 
131. Ducharme S, Price BH, Larvie M, Dougherty DD, Dickerson BCJAJoP. Clinical approach to the differential diagnosis between behavioral variant frontotemporal dementia and primary psychiatric disorders. 2015;172(9):827-37.
https://doi.org/10.1176/appi.ajp.2015.14101248
PMid:26324301
 
132. Wu S, Chai X, DeMattos R, Hutton M, Hayashi MJAs, Dementia. P4-314: A vast difference in the levels of insoluble tau in commonly used tau mouse models. 2013;9:P819-P.
https://doi.org/10.1016/j.jalz.2013.05.1760
 
133. Fleifel D, Rahmoon MA, AlOkda A, Nasr M, Elserafy M, El-Khamisy SFJJoGE, et al. Recent advances in stem cells therapy: A focus on cancer, Parkinson's and Alzheimer's. 2018;16(2):427-32.
https://doi.org/10.1016/j.jgeb.2018.09.002
PMid:30733756 PMCid:PMC6354001
 
134. Rascovsky K, Hodges JR, Knopman D, Mendez MF, Kramer JH, Neuhaus J, et al. Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia. 2011;134(9):2456-77.
 
135. Bang J, Spina S, Miller BLJTL. Frontotemporal dementia. 2015;386(10004):1672-82.
https://doi.org/10.1016/S0140-6736(15)00461-4
PMid:26595641
 
136. Porterfield V, Khan SS, Foff EP, Koseoglu MM, Blanco IK, Jayaraman S, et al. A three-dimensional dementia model reveals spontaneous cell cycle re-entry and a senescence-associated secretory phenotype. 2020;90:125-34.
https://doi.org/10.1016/j.neurobiolaging.2020.02.011
PMid:32184029 PMCid:PMC7166179
 
137. Ma H, Huang Y, Cong Z, Wang Y, Jiang W, Gao S, et al. The efficacy and safety of atypical antipsychotics for the treatment of dementia: a meta-analysis of randomized placebo-controlled trials. 2014;42(3):915-37.
https://doi.org/10.3233/JAD-140579
PMid:25024323
 
138. O'Brien JT, Holmes C, Jones M, Jones R, Livingston G, McKeith I, et al. Clinical practice with anti-dementia drugs: A revised (third) consensus statement from the British Association for Psychopharmacology. 2017;31(2):147-68.
https://doi.org/10.1177/0269881116680924
PMid:28103749
 
139. Logroscino G, Imbimbo BP, Lozupone M, Sardone R, Capozzo R, Battista P, et al. Promising therapies for the treatment of frontotemporal dementia clinical phenotypes: from symptomatic to disease-modifying drugs. 2019;20(9):1091-107.
https://doi.org/10.1080/14656566.2019.1598377
PMid:31002267
 
140. Mendez MFJDiCP. Frontotemporal dementia: therapeutic interventions. 2009;24:168-78.
https://doi.org/10.1159/000197896
PMid:19182475
 
141. Hallmann A-L, Araúzo-Bravo MJ, Mavrommatis L, Ehrlich M, Röpke A, Brockhaus J, et al. Astrocyte pathology in a human neural stem cell model of frontotemporal dementia caused by mutant TAU protein. 2017;7(1):42991.
https://doi.org/10.1038/srep42991
PMid:28256506 PMCid:PMC5335603
 
142. Tiwari S, Khan S, Kumar SV, Rajak R, Sultana A, Pasha SA, et al. Efficacy and safety of neural stem cell therapy for spinal cord injury: A systematic literature review. 2021;76(3):201-10.
https://doi.org/10.1016/j.therap.2020.06.011
PMid:32709426
 
143. Sivandzade F, Cucullo LJIJoMS. Regenerative stem cell therapy for neurodegenerative diseases: an overview. 2021;22(4):2153.
https://doi.org/10.3390/ijms22042153
PMid:33671500 PMCid:PMC7926761
 
144. Hautbergue GM, Cleary JD, Guo S, Ranum LPJCOiN. Therapeutic strategies for C9orf72 amyotrophic lateral sclerosis and frontotemporal dementia. 2021;34(5):748-55.
https://doi.org/10.1097/WCO.0000000000000984
PMid:34392299 PMCid:PMC8678157
 
145. Topiwala H, Terrera GM, Stirland L, Saunderson K, Russ TC, Dozier MF, et al. Lifestyle and neurodegeneration in midlife as expressed on functional magnetic resonance imaging: A systematic review. 2018;4:182-94.
https://doi.org/10.1016/j.trci.2018.04.001
PMid:29955662 PMCid:PMC6021545
 
146. Lewis PA, Spillane JE. The molecular and clinical pathology of neurodegenerative disease: Academic Press; 2018.
 
147. Mackenzie GJC-JdrdF. Will R. 2017;6(2053):10.12688.
https://doi.org/10.12688/f1000research.12681.1
PMid:29225787 PMCid:PMC5710312
 
148. Uttley L, Carroll C, Wong R, Hilton DA, Stevenson MJTLID. Creutzfeldt-Jakob disease: a systematic review of global incidence, prevalence, infectivity, and incubation. 2020;20(1):e2-e10.
https://doi.org/10.1016/S1473-3099(19)30615-2
PMid:31876504
 
149. Le N, Mercer R, Gojanovich AD, Park S, Wu B, Mostoslavsky G, et al. An iPSC library from a family with Creutzfeldt-Jakob disease caused by the E200K mutation. 2022.
 
150. Thody SA, Mathew M, Udgaonkar JBJBeBA-B. Mechanism of aggregation and membrane interactions of mammalian prion protein. 2018;1860(9):1927-35.
https://doi.org/10.1016/j.bbamem.2018.02.031
PMid:29514050
 
151. Sikorska B, Knight R, Ironside JW, Liberski PPJND. Creutzfeldt-Jakob disease. 2012:76-90.
https://doi.org/10.1007/978-1-4614-0653-2_6
PMid:22411235
 
152. Cobo F, Talavera P, Concha AJV. Diagnostic approaches for viruses and prions in stem cell banks. 2006;347(1):1-10.
https://doi.org/10.1016/j.virol.2005.11.026
PMid:16380145 PMCid:PMC7118799
 
153. Zambrano K, Barba D, Castillo K, Robayo P, Arizaga E, Caicedo A, et al. A new hope: Mitochondria, a critical factor in the war against prions. 2022.
https://doi.org/10.1016/j.mito.2022.05.004
PMid:35623560
 
154. Jensen JL, Berry DA, Kummer TAJPo. Investigating the effects of exam length on performance and cognitive fatigue. 2013;8(8):e70270.
https://doi.org/10.1371/journal.pone.0070270
PMid:23950918 PMCid:PMC3741288
 
155. Prusiner SBJSA. The prion diseases. 1995;272(1):48-57.
https://doi.org/10.1038/scientificamerican0195-48
PMid:7824915
 
156. Tagliavini F, McArthur R, Canciani B, Giaccone G, Porro M, Bugiani M, et al. Effectiveness of anthracycline against experimental prion disease in Syrian hamsters. 1997;276(5315):1119-21.
https://doi.org/10.1126/science.276.5315.1119
PMid:9148807
Volume 2, Issue 2
July 2023
Pages 55-68
  • Receive Date: 21 May 2023
  • Revise Date: 31 May 2023
  • Accept Date: 02 June 2023