Skip to main content
Ibrain logoLink to Ibrain
. 2024 Mar 12;10(1):93–105. doi: 10.1002/ibra.12152

Role and limitation of cell therapy in treating neurological diseases

Yu‐Qi Li 1, Peng‐Fei Li 1, Qian Tao 1, Issam J A Abuqeis 2, Yan‐Bin Xiyang 2,3,
PMCID: PMC11045202  PMID: 38682022

Abstract

The central role of the brain in governing systemic functions within human physiology underscores its paramount significance as the focal point of physiological regulation. The brain, a highly sophisticated organ, orchestrates a diverse array of physiological processes encompassing motor control, sensory perception, cognition, emotion, and the regulation of vital functions, such as heartbeat, respiration, and hormonal equilibrium. A notable attribute of neurological diseases manifests as the depletion of neurons and the occurrence of tissue necrosis subsequent to injury. The transplantation of neural stem cells (NSCs) into the brain exhibits the potential for the replacement of lost neurons and the reconstruction of neural circuits. Furthermore, the transplantation of other types of cells in alternative locations can secrete nutritional factors that indirectly contribute to the restoration of nervous system equilibrium and the mitigation of neural inflammation. This review summarized a comprehensive investigation into the role of NSCs, hematopoietic stem cells, mesenchymal stem cells, and support cells like astrocytes and microglia in alleviating neurological deficits after cell infusion. Moreover, a thorough assessment was undertaken to discuss extant constraints in cellular transplantation therapies, concurrently delineating indispensable model‐based methodologies, specifically on organoids, which were essential for guiding prospective research initiatives in this specialized field.

Keywords: cell therapy, mechanism, neurological diseases, organoids


Different types of cell xenotransplantation for treating neuro‐related diseases. Mesenchymal stem cells, neural stem cells, microglia, astrocytes, and hematopoietic stem cells are used to treat various neurological‐related diseases, including A β accumulation, neuronal death, brain atrophy, traumatic brain injury, nerve fiber entanglement, and so forth.

graphic file with name IBRA-10-93-g002.jpg

1. INTRODUCTION

The pathogenesis of neurological diseases is complex, involving multiple factors such as genetic mutations and the aging process. 1 , 2 Taking neurodegenerative diseases as an example, after the onset of the disease, significant characteristics include the loss of neurons, deficiencies in neurotrophic support, alterations in the neural microenvironment, localized tissue necrosis from injury, and increased gliosis. 3 , 4 If it can promote the regeneration of neurons and the reconstruction of neural circuits, it may promote the recovery of neurodegenerative diseases. 5 Numerous studies have shown that cell therapy is effective for neurodegenerative diseases. 6 As a consequence, the utilization of cell transplantation has emerged as an auspicious therapeutic modality. 7 Neural stem cells (NSCs) possess the inherent capability to undergo differentiation into a spectrum of cell types, comprising astrocytes, oligodendrocytes, and neurons. 2 , 8 A wealth of research findings indicated that subsequent to NSC transplantation, a substantial degree of integration with host cells ensues, thereby facilitating the reconstruction of neural circuits and the restoration of motor and cognitive functions, as substantiated by observations in animal models. 5 , 9 Additionally, mesenchymal stem cells (MSCs) have strong immunomodulatory capabilities and secretion functions, allowing them to provide nourishment to locally injured tissues and effectively regulate the inflammatory microenvironment. 10 , 11 As the aging process unfolds, the manifestation of an inflammatory microenvironment assumes noteworthy prominence. The subtle intricacies of an immune microenvironment can instigate the activation of microglia, designating these cellular entities as a primary locus for addressing therapeutic interventions in the context of neurodegenerative diseases. 12 , 13 In conditions characterized by abnormal microglia activation, strategies aimed at suppressing this irregularity. Hematopoietic stem cells (HSCs) can migrate to the brain and develop into microglia, replacing host microglia through exogenous transplantation, and establishing transplanted HSCs within the brain to reconstruct its microenvironment are crucial in neurodegenerative disease therapeutics. 14 Astrocytes are the most widely distributed type of cells in the brain, located between the cell body and its processes of nerve cells, supporting and guiding neurons, enhancing their survival, and promoting synaptic connections between neurons. 15 , 16 , 17 Similarly, astrocytes play a multifaceted role in the brain, undertaking various functions, such as supplying nourishment, reinforcing neuronal structure, and facilitating the clearance of debris, making them potential to treat neurological diseases. 18 Together, this review summarized the role of NSCs, MSCs, astrocytes, microglia, and HSCs in the treatment of neurological diseases through transplantation. Furthermore, this study conducts a critical assessment of the existing limitations inherent in current transplantation methods and delves into potential avenues like organoids for optimizing therapeutic strategies.

2. MECHANISMS OF HETEROGENEOUS CELL TRANSPLANTATION FOR NEUROLOGICAL DISEASE THERAPY

2.1. Transplantation of NSCs for neurological diseases

Neurodegenerative diseases, colloquially termed the “silent killers” within the geriatric demographic, predominantly involve the loss of neurons, a phenomenon further compounded by the heightened susceptibility inherent in mature nervous systems. 4 , 19 NSCs, residing within the central nervous system, possess the capacity to generate diverse neural components, encompassing neurons, astrocytes, and oligodendrocytes. 16 , 20 Predominantly localized in regions, such as the subventricular zone, dentate gyrus of the hippocampus, and the hypothalamus, NSCs assume a pivotal role in the therapeutic management of neurodegenerative diseases, 21 , 22 , 23 and manifest the capability to undergo differentiation into a spectrum of cell types, including neurons, astrocytes, and oligodendrocytes. 24 The consequential roles played by these differentiated cell types are crucial, encompassing the replacement of lost neurons, provision of structural support, and facilitation of the repair of damaged myelin 25 , 26 (Figure 1). Neurodegenerative and traumatic diseases affecting the nervous system, such as axonal malnutrition, 27 Parkinson's disease, 28 stroke, 29 brain injury, 30 and spinal cord injury, 31 , 32 , 33 lead to tissue necrosis and physiological damage in brain tissue. However, the endogenous production of NSCs significantly diminishes as the nervous system matures, thereby restricting their potential to effectively repair neural damage caused by these conditions. Consequently, supplementing lost neurons or stimulating their regeneration typically necessitates an external source.

Figure 1.

Figure 1

NSCs differentiate into neurons to supplement the neurons lost in the brain due to diseases, oligodendrocytes to encapsulate neurons and prevent abnormal neuronal discharge, and astrocytes, providing both nutrition and structural support for neurons. NSC, neural stem cell. [Color figure can be viewed at wileyonlinelibrary.com]

A multitude of studies underscore the profound therapeutic impacts of NSCs on various ailments, such as Parkinson's and Huntington's diseases. Transplanted NSCs not only assimilate into the recipient's brain, establishing novel synaptic connections but also secrete neurotrophic factors that modulate immune responses and confer neuroprotective properties. 34 , 35 , 36 As a result, allogeneic cell transplantation has emerged as a viable approach to address physical brain injuries and physiological abnormalities. Studies have shown that NSCs can provide nourishment to damaged cells and regulate immune and metabolic pathways through a bystander effect. 37 , 38 , 39 These NSCs are initially obtained from the telencephalon and diencephalon of human fetal brains after selective pregnancy terminations between 10 and 12 weeks postconception. 40 Before transplantation into patients, the NSCs undergo in vitro expansion, quality control, and safety assessments at different dosage levels. 41 The experimental findings furnish evidence that the transplantation of NSCs is associated with a reduction in overall brain and gray matter volume loss. 42 Furthermore, the examination of untargeted proteomics and metabolomics in the cerebrospinal fluid of subjects subjected to NSC transplantation indicates that these cells operate as neuroprotective agents, functioning in nutrient provision and immune system regulation. 43

2.2. MSC transplantation for neurological diseases

MSCs possess a remarkable capacity to differentiate into a wide array of cell types, including osteoblasts, chondrocytes, adipocytes, bone marrow stromal cells, and liver cells. Consequently, they hold significant promise in promoting neural regeneration following injury. 44 , 45 These MSCs can be obtained from various sources, such as umbilical cord blood, umbilical cords, amniotic membranes and fluid, bone marrow, and placenta, and are characterized by their limited immunogenicity. The extensive accessibility, expandability, minimal immunogenicity, and notable therapeutic efficacy of MSCs make them a prominent option for cell‐based therapies aimed at neurorelated disorders. MSCs possess a diverse range of capabilities, encompassing immunomodulation, angiogenesis, tissue remodeling, antiapoptosis, growth factor secretion, and nourishment (Figure 2). 46 , 47 , 48 Collectively, these attributes facilitate the survival of host cells, tissue restoration, and the stimulation or differentiation of local progenitor cells. 49

Figure 2.

Figure 2

The mechanism involved in MSC transplantation for treating neurological diseases. MSCs release anti‐inflammatory factors to reduce abnormal activation of microglia and restore them to a resting state. They provide nutritional factors for astrocytes and neurons to reduce neuronal death. MSC, mesenchymal stem cell. [Color figure can be viewed at wileyonlinelibrary.com]

Additionally, the extracellular vesicles derived from MSCs during stroke exhibit a remarkable ability to selectively migrate toward sites of injury, thereby safeguarding neural tissues against additional necrotic expansion, promoting late‐stage vascularization, and releasing growth factors that facilitate tissue remodeling. 49 In the context of cotransplantation with other cell types, MSCs exhibit the capability to stimulate the differentiation of NSCs and augment the survival rates of transplanted NSCs. 50 , 51 These distinctive attributes position MSCs as a promising therapeutic modality for mitigating inflammation and addressing neurodegenerative alterations linked to various neuro‐related diseases across diverse clinical scenarios.

2.3. Transplantation of astrocytes to treat neurological diseases

Astrocytes, derived from the neural epithelium, assume a crucial role in upholding homeostasis in the central nervous system. 15 , 16 They exhibit a wide array of functions, encompassing molecular to systemic domains, such as regulating ion equilibrium, providing metabolic support, modulating synaptic connections, safeguarding the blood–brain barrier, and clearing cellular debris 52 , 53 (Figure 3). Empirical evidence indicates that cotransplantation of NSCs and astrocytes yields substantial improvements in neural functionality in rodent models of ischemic stroke, surpassing the outcomes achieved through individual NSC transplantation. 54 , 55 Furthermore, this integrated approach notably mitigates the extent of brain infarction volume when contrasted with the sole transplantation of NSCs. 56 Research findings indicate that the transplantation of mature neural glial progenitors in the adult brain facilitates the reorganization of the sensory cortex through their functional integration into the pre‐existing astrocyte–neuron circuitry. 57 Furthermore, the introduction of glial precursor cells (GPCs) into the cortex of adult mice leads to the development of astrocytes with typical functional attributes following in vitro cultivation. These transplanted cells demonstrate remarkable abilities in terms of migration, differentiation, and long‐term integration, thereby preserving a more youthful state within the aging brain. Significantly, the transplantation of GPCs has demonstrated the ability to reverse age‐related sensory sensitivity. 58 After spinal cord injury, astrocytes can promote the regeneration of damaged axons and the sprouting of undamaged axons by secreting appropriate extracellular matrix and neurotrophic factors, participating in immune regulation, and suppressing inflammatory responses. Studies have shown that transplantation of astrocyte lines can enhance axonal regeneration. 59 The burgeoning body of empirical evidence emphasizes the potential efficacy of astrocyte transplantation as a promising therapeutic approach in the treatment of neurological disorders. This is particularly noteworthy for its impact on the modulation of neural circuitry and the rejuvenation of brain function.

Figure 3.

Figure 3

The mechanism involved in astrocyte transplantation for treating neurological diseases. Astrocytes provide nutrients and structural support to neurons and rescue them from death. [Color figure can be viewed at wileyonlinelibrary.com]

2.4. The therapeutic potential of transplanted microglia in neurological diseases

Microglia, highly prevalent in cerebral tissue, are posited to have mesodermal origins and fulfill indispensable roles within the central nervous system. 12 Their significant contributions encompass participation in brain development, maintenance of homeostasis, responsiveness to neural inflammation, and active engagement in aging processes. 60 , 61 , 62 Particularly during early brain development, microglia actively participate in synaptic pruning, a vital process responsible for the elimination of excess synapses. 63 However, in neurodegenerative disorders, such as advanced Alzheimer's disease, the aggregation of β‐amyloid proteins within the brain results in neurotoxic consequences, causing harm to neurons and triggering the activation of microglia. 64 , 65 Microglia demonstrate a range of functional phenotypes in response to various conditions. Following neural injury, they initially assume an activated M1 phenotype to facilitate the removal of cellular debris. 66 Subsequently, they transition into an anti‐inflammatory M2 phenotype, releasing cytokines, such as interleukin‐4 (IL‐4) and IL‐10, thereby contributing to the mitigation of inflammation. 67 , 68

Owing to their functions in surveillance and clearance processes, microglia have become a focal point in scientific investigations aimed at addressing related pathological conditions. 69 Research demonstrated that the inhibition of microglia triggers their swift repopulation within the brain, resulting in the emergence of neuroprotective microglia 70 , 71 , 72 (Figure 4). The efficacy of these regenerated microglia is contingent upon the signaling of IL‐6 via the soluble IL‐6 receptor, which facilitates adult neurogenesis and augments the viability of nascent neurons essential for cognitive processes. 73 Significant advancements in the healing process and regeneration of axons within the spinal cord, a vital constituent of the central nervous system, have been documented through therapeutic interventions encompassing the transplantation of neonatal microglia or adult microglia treated with peptidase inhibitors. 12 , 74 , 75 Nevertheless, the seamless integration of externally transplanted microglia into the recipient's brain continues to present a formidable obstacle. In 2020, three discrete techniques have been devised for the transplantation of microglia, each demonstrating efficacy in successfully engrafting the transplanted cells into the brain. 76

Figure 4.

Figure 4

Mechanisms of transplanted microglia in addressing neurological diseases. The drug inhibits abnormal activation of microglia in the host's brain, while brain transplantation supplements healthy microglia. Microglia maintain brain homeostasis after self‐renewal. [Color figure can be viewed at wileyonlinelibrary.com]

2.5. HSC transplantation in neurological disease treatment

HSCs are a cohort of undifferentiated cells located within hematopoietic tissues, responsible for generating all blood and immune cells. 77 , 78 These cells possess the distinctive ability to undergo specialization into fully developed red blood cells, white blood cells, and platelets, while simultaneously preserving the vital functions of self‐renewal and replication, which are imperative for regular hematopoiesis and immune response. 79 , 80 Recent investigations suggest that subsequent to transplantation, HSCs can differentiate into microglial cells and establish a presence within the cerebral region 81 , 82 (Figure 5). Research findings indicate that transplants obtained from HSCs and progenitor cells effectively mitigate various symptoms and manifestations in murine models of Alzheimer's disease. 14 , 83 In contrast to other mice affected by Alzheimer's, those who receive HSC transplants exhibit maintained memory and cognitive abilities, diminished neural inflammation, and a noteworthy reduction in the accumulation of β‐amyloid protein. 84 Significantly, the research conducted by Marius Wernig and his team revealed that the systemic transplantation of HSCs has the ability to replace mutated microglia in triggering receptor expressed on myeloid cells 2‐mutant mice models of Alzheimer's disease, effectively restoring their functionality. These findings provide compelling evidence that HSC transplantation exhibits substantial potential as a prospective therapeutic strategy for forthcoming treatments of Alzheimer's disease. 14

Figure 5.

Figure 5

HSCs transplantation in neurological disease treatment. After HSCs transplantation, differentiation into microglia can migrate and colonize the brain to maintain neural homeostasis. HSC, hematopoietic stem cell. [Color figure can be viewed at wileyonlinelibrary.com]

3. LIMITING FACTORS IN HETEROGENEOUS CELL TRANSPLANTATION FOR TREATING NEUROLOGICAL DISEASES

3.1. Immune rejection after cell transplantation

The field of organ and cell transplantation has made significant progress in the treatment of diverse medical conditions. 85 , 86 , 87 Nevertheless, the pervasive challenge of immune rejection remains a significant impediment across all transplant procedures, ultimately contributing to instances of transplant failures. 88 , 89 , 90 Currently, the management of immune rejection heavily relies on the administration of immunosuppressants, including cyclosporine, tacrolimus, and dexamethasone, with the primary objective of preventing rejection. 91 , 92 , 93 However, prolonged usage of these medications frequently compromises the recipient's immune system, leading to infections and inadequate suppression of transplanted cells. 94 , 95 , 96 According to research findings, regulatory T cells have been found to exert a temporary inhibitory effect on the proliferation of CD8+ T cells, thereby providing a limited defense against rejection for a duration of approximately 2 months. 97 However, the unresolved matter of persistent immune rejection continues to be a matter of great concern. Specifically, in the context of intracranial cell transplantation for neurological disorders, the principal factor contributing to cell demise at the transplantation site is the substantial infiltration of T cells, B cells, and natural killer cells originating from blood vessels and peripheral tissues, which subsequently triggers immune rejection and self‐inflammation. 98 , 99 Dendritic cells (DCs) are the most functional specialized antigen‐presenting cells in the body. 100 , 101 In addition to playing an important role in the body's innate immune response, they are also responsible for initiating adaptive immune responses and influencing the types of immune responses, assisting in the development of immune effects. 102 The surface of the DC membrane contains abundant major histocompatibility complex (MHC) class I and II, costimulatory molecules (CD80/B7‐1, CD86/B7‐2, CD40, CD40 ligands, etc.), intercellular adhesion molecule‐1 (ICAM‐1), ICAM‐2, ICAM‐3, and lymphocyte function related molecule‐1, which can stimulate initial T‐cell activation and proliferation. 103 , 104 The reactivation of microglia through infiltration of peripheral immune cells results in an upregulation of MHC class II, resembling that of peripheral DCs. 105 This increased expression facilitates the presentation of antigen signals to infiltrating T cells, thereby establishing a connection between the immune systems of the peripheral and central nervous systems. 106 This phenomenon persists until the restoration of the blood‐brain barrier, ultimately leading to the demise of transplanted cells originating from an external source. 107 , 108

3.2. Divergent brain development across species

Early‐stage transplantation experiments involving human‐origin cells are restricted to rodent and primate models as recipients, primarily due to ethical and moral considerations. 109 , 110 The existence of developmental disparities among primate, rodent, and human brains poses significant obstacles in establishing long‐lasting neural connections during interspecies cell transplantation. 111 , 112 , 113 Notably, the brain stands out as a prominent differentiating factor between humans and animals. 114 , 115 , 116 The human brain, which is approximately three times larger than that of chimpanzees, accommodates a greater number of neurons and glial cells, leading to an augmentation in neural pathways and the emergence of specialized functional regions, specifically associated with distinct human cognitive capabilities. 117 , 118 , 119 The evolutionary mechanisms have exerted an influence on the initial formation of neurons in both chimpanzees and our human predecessors. In comparison to humans, the chimpanzee genome exhibits a higher abundance of Line 1 (L1) elements that are specific to the species, suggesting a heightened rate of L1 migration observed in non‐human primates beyond cultured induced pluripotent stem cells (iPSCs). 120 The alterations in brain gene transcription over temporal and spatial dimensions exert an impact on the molecular, morphological, and functional characteristics of the brain. A study undertaken by the research team, led by J. Gray Camp encompasses the utilization of single‐cell transcriptomics and chromatin studies, spanning from early pluripotent cells such as embryonic stem cells and iPSCs to organoid structures, across various primate species, including humans, chimpanzees, and monkeys. 121 Hence, a significant challenge in investigating the transplantation of human‐origin cells into different species lies in the incongruent developmental stages observed among these species.

4. FUTURE STRATEGIES FOR TREATING NEUROLOGICAL DISEASES

4.1. Brain organoid substitutes for animal models in neurological disease

Organoids are three‐dimensional structures that are created in a laboratory setting using pluripotent stem cells (PSCs) or adult stem cells. 122 , 123 These structures have the ability to self‐organize and mimic the cell compositions, tissue architectures, and functionalities of specific organs. 124 The development of organoids was initially driven by advancements in human PSC technologies. 125 Moscona, 126 accomplished a noteworthy breakthrough in this domain and demonstrated the formation of organoid‐like structures through dissociation–aggregation experiments on embryonic neural tissue, resembling their in vivo counterparts. Zhang et al., 127 developed methods to direct the differentiation of human PSCs toward neural epithelial cells, enabling the creation of basic two‐dimensional models that mimic different cortical structures in the human brain. However, the crucial advancement toward the creation of the first human brain organoid occurred with the transition to three‐dimensional models. Nakano et al., 128 spearheaded this shift by cultivating PSCs‐derived neural epithelial cells in a fully three‐dimensional setting, guiding their maturation into optic nerve discs. This milestone played a pivotal role in establishing the foundation for further progress in retinal organoid development. Neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease, are primarily sporadic and intricate in their etiology. 129 , 130 The complex biological underpinnings of conditions like depression, autism spectrum disorders, and schizophrenia present challenges in replicating them in rodent models and assessing treatment effectiveness. Generally, non‐human primate models, which closely resemble human behaviors, are limited by high cost and rare sample availability for high‐throughput screenings. However, brain organoids cultivated in vitro offer a cost‐effective and efficient means to investigate the intricate mechanisms underlying these diseases.

4.2. Transplantation of in vitro‐cultured brain‐organoid tissues for neurological diseases

Understanding the development and dysfunction of the human brain is a major goal of neurobiology. The brain organoids created by human PSCs represent a promising method for brain repair. 131 They acquire many structural features of the brain and increase the likelihood of patient matching and repair. 132 A recent notable advancement has been made by scientists who successfully transplanted brain tissue organoids derived from human cells into mice suffering from stroke. 133 This transplantation procedure effectively restored impaired tissue and reinstated motor functions. 134 , 135 Furthermore, upon transplantation into the visual cortex of rat brains, the brain organoids exhibited seamless integration into the host tissue, exhibiting progressive development of blood vessels, enlargement in size, generation of neuronal projections, and establishment of synaptic connections. 136 , 137 Moreover, the introduction of human brain organoids into the brains of neonatal rats, specifically those aged 2–3 days, a critical period characterized by ongoing neural connectivity development, resulted in the subsequent growth and occupation of approximately one‐third of the rat brain hemisphere by these transplanted human brain organoids. 132 Notably, the neurons within these organoids successfully established functional connections with the existing neural circuitry, leading to the activation of these transplanted neural components and subsequent restoration of specific functionalities within the corresponding brain regions. 138

5. CONCLUSIONS AND PROSPECTS

The brain, constituting a sophisticated system, integrates distinct cell populations to collectively execute multifaceted functions. Pathological conditions within the brain encompass a spectrum of underlying mechanisms, including neuronal malnutrition, extensive neuronal death, gene mutations, and glial cell activation. 139 These mechanisms necessitate the exploration and development of diverse cell transplantation techniques. Cell transplantation, including astrocytes, microglia, and NSCs, has the potential to mitigate neuronal malnutrition, holding promise in ameliorating the inflammatory milieu within the brain. 17 In addition, HSC transplantation facilitates the reconstruction of the hematopoietic system, wherein certain stem cells undergo differentiation into oligodendrocytes within the cerebral region. 14 The transplantation of MSCs has been shown to mitigate brain inflammation by promoting neuronal growth and reinstating the brain's homeostasis via nutrient secretion. 140 Nevertheless, the principal constraint of cell transplantation presently resides in immune suppression, thereby demanding the investigation of innovative immune‐suppressive approaches, combinations of diverse immune‐suppressive agents, and the minimization of their utilization. 141 , 142

As a result of ethical considerations, cells cultivated in vitro are limited to initial validation in animal models. 143 Non‐human primates, which demonstrate phenotypes more closely resembling those of humans, are favored in disease animal model investigations; however, they encounter obstacles associated with costs. Brain organoids provide a practical avenue for the examination of genetic mutations or diseases associated with developmental anomalies. 144 Serving as valuable research models, these organoids can be successfully engrafted into the host brain through transplantation. 132 Nonetheless, it is imperative to acknowledge that brain organoids are subject to specific limitations arising from their enlarged size and volume, rendering them more applicable to scenarios involving surface damage in the cerebral cortex and comparable circumstances.

AUTHOR CONTRIBUTIONS

Yu‐Qi Li wrote the original manuscript and drew the figures. Qian Tao collected literature. Peng‐Fei Li, Issam J. A. Abuqeis, and Yan‐Bin Xiyang fine‐tuned the whole article. Yan‐Bin Xiyang finalized and approved this paper. All authors have read and approved the final submitted manuscript.

CONFLICT OF INTEREST STATEMENT

The authors declare no conflicts of interest.

ETHICS STATEMENT

Not applicable.

ACKNOWLEDGMENTS

Not applicable. The author declares funding as none.

Li Y‐Q, Li P‐F, Tao Q, Abuqeis IJA, Xiyang Y‐B. Role and limitation of cell therapy in treating neurological diseases. ibrain. 2024;10:93‐105. 10.1002/ibra.12152

Yu‐Qi Li and Peng‐Fei Li contributed equally.

DATA AVAILABILITY STATEMENT

Not applicable.

REFERENCES

  • 1. López‐Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023;186(2):243‐278. 10.1016/j.cell.2022.11.001 [DOI] [PubMed] [Google Scholar]
  • 2. Chi H, Chang H‐Y, Sang T‐K. Neuronal cell death mechanisms in major neurodegenerative diseases. Int J Mol Sci. 2018;19(10):3082. 10.3390/ijms19103082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. De Gioia R, Biella F, Citterio G, et al. Neural stem cell transplantation for neurodegenerative diseases. Int J Mol Sci. 2020;21(9):3103. 10.3390/ijms21093103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Checkoway H, Lundin JI, Kelada SN. Neurodegenerative diseases. IARC Sci Publ. 2011;(163):407‐419. [PubMed] [Google Scholar]
  • 5. Temple S. Advancing cell therapy for neurodegenerative diseases. Cell Stem Cell. 2023;30(5):512‐529. 10.1016/j.stem.2023.03.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Lunn JS, Sakowski SA, Hur J, Feldman EL. Stem cell technology for neurodegenerative diseases. Ann Neurol. 2011;70(3):353‐361. 10.1002/ana.22487 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Dantuma E, Merchant S, Sugaya K. Stem cells for the treatment of neurodegenerative diseases. Stem Cell Res Ther. 2010;1(5):37. 10.1186/scrt37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Huang L, Zhang L. Neural stem cell therapies and hypoxic‐ischemic brain injury. Prog Neurobiol. 2019;173:1‐17. 10.1016/j.pneurobio.2018.05.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Rumajogee P, Altamentova S, Li L, et al. Exogenous neural precursor cell transplantation results in structural and functional recovery in a hypoxic–ischemic hemiplegic mouse model. eNeuro. 2018;5(5):eENEURO.0369‐18.2018. 10.1523/ENEURO.0369-18.2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Du L, Lin L, Li Q, et al. IGF‐2 preprograms maturing macrophages to acquire oxidative phosphorylation‐dependent anti‐inflammatory properties. Cell Metab. 2019;29(6):1363‐1375. 10.1016/j.cmet.2019.01.006 [DOI] [PubMed] [Google Scholar]
  • 11. Zhou H, He Y, Xiong W, et al. MSC based gene delivery methods and strategies improve the therapeutic efficacy of neurological diseases. Bioact Mater. 2023;23:409‐437. 10.1016/j.bioactmat.2022.11.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Prinz M, Jung S, Priller J. Microglia biology: one century of evolving concepts. Cell. 2019;179(2):292‐311. 10.1016/j.cell.2019.08.053 [DOI] [PubMed] [Google Scholar]
  • 13. Chen Z, Trapp BD. Microglia and neuroprotection. J Neurochem. 2016;136(suppl 1):10‐17. 10.1111/jnc.13062 [DOI] [PubMed] [Google Scholar]
  • 14. Yoo Y, Neumayer G, Shibuya Y, Mader MM‐D, Wernig M. A cell therapy approach to restore microglial Trem2 function in a mouse model of Alzheimer's disease. Cell Stem Cell. 2023;30(8):1043‐1053.e6. 10.1016/j.stem.2023.07.006 [DOI] [PubMed] [Google Scholar]
  • 15. Endo F, Kasai A, Soto JS, et al. Molecular basis of astrocyte diversity and morphology across the CNS in health and disease. Science. 2022;378(6619):eadc9020. 10.1126/science.adc9020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Zhou B, Zuo Y‐X, Jiang R‐T. Astrocyte morphology: diversity, plasticity, and role in neurological diseases. CNS Neurosci Ther. 2019;25(6):665‐673. 10.1111/cns.13123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Yang Y, Song J‐J, Choi YR, et al. Therapeutic functions of astrocytes to treat α‐synuclein pathology in Parkinson's disease. Proc Natl Acad Sci USA. 2022;119(29):e2110746119. 10.1073/pnas.2110746119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Lawal O, Ulloa Severino FP, Eroglu C. The role of astrocyte structural plasticity in regulating neural circuit function and behavior. GLIA. 2022;70(8):1467‐1483. 10.1002/glia.24191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Dugger BN, Dickson DW. Pathology of neurodegenerative diseases. Cold Spring Harbor Perspect Biol. 2017;9(7):a028035. 10.1101/cshperspect.a028035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Guo N, McDermott KD, Shih Y‐T, et al. Transcriptional regulation of neural stem cell expansion in the adult hippocampus. eLife. 2022;11:e72195. 10.7554/eLife.72195 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Gonçalves JT, Schafer ST, Gage FH. Adult neurogenesis in the hippocampus: from stem cells to behavior. Cell. 2016;167(4):897‐914. 10.1016/j.cell.2016.10.021 [DOI] [PubMed] [Google Scholar]
  • 22. Li Y‐D, Luo Y‐J, Chen Z‐K, et al. Hypothalamic modulation of adult hippocampal neurogenesis in mice confers activity‐dependent regulation of memory and anxiety‐like behavior. Nat Neurosci. 2022;25(5):630‐645. 10.1038/s41593-022-01065-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Makrygianni EA, Chrousos GP. Neural progenitor cells and the hypothalamus. Cells. 2023;12(14):1822. 10.3390/cells12141822 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Tuazon JP, Castelli V, Lee J‐Y, et al. Neural stem cells. Adv Exp Med Biol. 2019;1201:79‐91. 10.1007/978-3-030-31206-0_4 [DOI] [PubMed] [Google Scholar]
  • 25. Willis CM, Nicaise AM, Peruzzotti‐Jametti L, Pluchino S. The neural stem cell secretome and its role in brain repair. Brain Res. 2020;1729: 146615. 10.1016/j.brainres.2019.146615 [DOI] [PubMed] [Google Scholar]
  • 26. Gaillard A, Prestoz L, Dumartin B, et al. Reestablishment of damaged adult motor pathways by grafted embryonic cortical neurons. Nat Neurosci. 2007;10(10):1294‐1299. 10.1038/nn1970 [DOI] [PubMed] [Google Scholar]
  • 27. Ma L, Hu B, Liu Y, et al. Human embryonic stem cell‐derived GABA neurons correct locomotion deficits in quinolinic acid‐lesioned mice. Cell Stem Cell. 2012;10(4):455‐464. 10.1016/j.stem.2012.01.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Moriarty N, Gantner CW, Hunt CPJ, et al. A combined cell and gene therapy approach for homotopic reconstruction of midbrain dopamine pathways using human pluripotent stem cells. Cell Stem Cell. 2022;29(3):434‐448. 10.1016/j.stem.2022.01.013 [DOI] [PubMed] [Google Scholar]
  • 29. Walker AE. Editorial: guidelines for stroke. Surg Neurol. 1974;2(2):108. [PubMed] [Google Scholar]
  • 30. Overgaard J, Tweed WA. Cerebral circulation after head injury. Part 2: the effects of traumatic brain edema. J Neurosurg. 1976;45(3):292‐300. 10.3171/jns.1976.45.3.0292 [DOI] [PubMed] [Google Scholar]
  • 31. Baloh RH, Johnson JP, Avalos P, et al. Transplantation of human neural progenitor cells secreting GDNF into the spinal cord of patients with ALS: a phase 1/2a trial. Nat Med. 2022;28(9):1813‐1822. 10.1038/s41591-022-01956-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Álvarez Z, Kolberg‐Edelbrock AN, Sasselli IR, et al. Bioactive scaffolds with enhanced supramolecular motion promote recovery from spinal cord injury. Science. 2021;374(6569):848‐856. 10.1126/science.abh3602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Tao Y, Vermilyea SC, Zammit M, et al. Autologous transplant therapy alleviates motor and depressive behaviors in parkinsonian monkeys. Nat Med. 2021;27(4):632‐639. 10.1038/s41591-021-01257-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Xiong M, Tao Y, Gao Q, et al. Human stem cell‐derived neurons repair circuits and restore neural function. Cell Stem Cell. 2021;28(1):112‐126. 10.1016/j.stem.2020.08.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Luo L. Architectures of neuronal circuits. Science. 2021;373(6559):1103. eabg7285 10.1126/science.abg7285 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Haney MJ, Klyachko NL, Zhao Y, et al. Exosomes as drug delivery vehicles for Parkinson's disease therapy. J Control Release. 2015;207:18‐30. 10.1016/j.jconrel.2015.03.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Butti E, Cattaneo S, Bacigaluppi M, et al. Neural precursor cells tune striatal connectivity through the release of IGFBPL1. Nat Commun. 2022;13(1):7579. 10.1038/s41467-022-35341-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Zhang R, Zhang Z, Chopp M. Function of neural stem cells in ischemic brain repair processes. J Cereb Blood Flow Metab. 2016;36(12):2034‐2043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Martín‐Suárez S, Valero J, Muro‐García T, Encinas JM. Phenotypical and functional heterogeneity of neural stem cells in the aged hippocampus. Aging cell. 2019;18(4):e12958. 10.1111/acel.12958 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Ferrari D, Gelati M, Profico DC, Vescovi AL. Human fetal neural stem cells for neurodegenerative disease treatment. Results Probl Cell Differ. 2018;66:307‐329. 10.1007/978-3-319-93485-3_14 [DOI] [PubMed] [Google Scholar]
  • 41. Fernandez‐Muñoz B, Garcia‐Delgado AB, Arribas‐Arribas B, Sanchez‐Pernaute R. Human neural stem cells for cell‐based medicinal products. Cells. 2021;10(9):2377. 10.3390/cells10092377 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Li Y, Tran A, Graham L, Brock J, Tuszynski MH, Lu P. BDNF guides neural stem cell‐derived axons to ventral interneurons and motor neurons after spinal cord injury. Exp Neurol. 2023;359:114259. 10.1016/j.expneurol.2022.114259 [DOI] [PubMed] [Google Scholar]
  • 43. Genchi A, Brambilla E, Sangalli F, et al. Neural stem cell transplantation in patients with progressive multiple sclerosis: an open‐label, phase 1 study. Nat Med. 2023;29(1):75‐85. 10.1038/s41591-022-02097-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Yousefi F, Lavi Arab F, Nikkhah K, Amiri H, Mahmoudi M. Novel approaches using mesenchymal stem cells for curing peripheral nerve injuries. Life Sci. 2019;221:99‐108. 10.1016/j.lfs.2019.01.052 [DOI] [PubMed] [Google Scholar]
  • 45. Bojanic C, To K, Zhang B, Mak C, Khan WS. Human umbilical cord derived mesenchymal stem cells in peripheral nerve regeneration. World J Stem Cells. 2020;12(4):288‐302. 10.4252/wjsc.v12.i4.288 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Secco M, Zucconi E, Vieira NM, et al. Multipotent stem cells from umbilical cord: cord is richer than blood!. Stem Cells. 2008;26(1):146‐150. 10.1634/stemcells.2007-0381 [DOI] [PubMed] [Google Scholar]
  • 47. Ding DC, Chang YH, Shyu WC, Lin SZ. Human umbilical cord mesenchymal stem cells: a new era for stem cell therapy. Cell Transplant. 2015;24(3):339‐347. 10.3727/096368915x686841 [DOI] [PubMed] [Google Scholar]
  • 48. Kubiak CA, Grochmal J, Kung TA, Cederna PS, Midha R, Kemp SWP. Stem‐cell‐based therapies to enhance peripheral nerve regeneration. Muscle Nerve. 2020;61(4):449‐459. 10.1002/mus.26760 [DOI] [PubMed] [Google Scholar]
  • 49. Myers MI, Hines KJ, Gray A, Spagnuolo G, Rosenwasser R, Iacovitti L. Intracerebral transplantation of autologous mesenchymal stem cells improves functional recovery in a rat model of chronic ischemic stroke. Transl Stroke Res. Published online November 2, 2023. 10.1007/s12975-023-01208-7 [DOI] [PMC free article] [PubMed]
  • 50. Campos HC, Ribeiro DE, Hashiguchi D, et al. Distinct effects of the hippocampal transplantation of neural and mesenchymal stem cells in a transgenic model of Alzheimer's disease. Stem Cell Rev Rep. 2022;18(2):781‐791. 10.1007/s12015-021-10321-9 [DOI] [PubMed] [Google Scholar]
  • 51. Ratushnyak MG, Semochkina YP, Zhirnik AS, Smirnova OD. Improved survival and regeneration of irradiated mouse neural stem cells after co‐culturing with non‐irradiated mouse neural stem cells or mesenchymal stem cells from the adipose tissue. Bull Exp Biol Med. 2021;172(2):228‐235. 10.1007/s10517-021-05368-0 [DOI] [PubMed] [Google Scholar]
  • 52. Stevens B. Neuron‐astrocyte signaling in the development and plasticity of neural circuits. Neurosignals. 2008;16(4):278‐288. 10.1159/000123038 [DOI] [PubMed] [Google Scholar]
  • 53. Zhou T, Li Y, Li X, et al. Microglial debris is cleared by astrocytes via C4b‐facilitated phagocytosis and degraded via RUBICON‐dependent noncanonical autophagy in mice. Nat Commun. 2022;13(1):6233. 10.1038/s41467-022-33932-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Lundberg C, Martínez‐Serrano A, Cattaneo E, McKay RDG, Björklund A. Survival, integration, and differentiation of neural stem cell lines after transplantation to the adult rat striatum. Exp Neurol. 1997;145(Pt 1):342‐360. [DOI] [PubMed] [Google Scholar]
  • 55. Moshayedi P, Nih LR, Llorente IL, et al. Systematic optimization of an engineered hydrogel allows for selective control of human neural stem cell survival and differentiation after transplantation in the stroke brain. Biomaterials. 2016;105:145‐155. 10.1016/j.biomaterials.2016.07.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Wen SJ, Zheng XM, Liu LF, et al. Effects of primary microglia and astrocytes on neural stem cells in in vitro and in vivo models of ischemic stroke. Neural Regen Res. 2021;16(9):1677‐1685. 10.4103/1673-5374.306093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Zhang K, Chen C, Yang Z, et al. Sensory esponse of transplanted astrocytes in adult mammalian cortex in vivo. Cereb Cortex. 2016;26(9):3690‐3704. 10.1093/cercor/bhw213 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Yang Z, Gong M, Jian T, et al. Engrafted glial progenitor cells yield long‐term integration and sensory improvement in aged mice. Stem Cell Res Ther. 2022;13(1):285. 10.1186/s13287-022-02959-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Goulão M, Ghosh B, Urban MW, et al. Astrocyte progenitor transplantation promotes regeneration of bulbospinal respiratory axons, recovery of diaphragm function, and a reduced macrophage response following cervical spinal cord injury. GLIA. 2019;67(3):452‐466. 10.1002/glia.23555 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Nayak D, Roth TL, McGavern DB. Microglia development and function. Annu Rev Immunol. 2014;32:367‐402. 10.1146/annurev-immunol-032713-120240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Colonna M, Butovsky O. Microglia function in the central nervous system during health and neurodegeneration. Annu Rev Immunol. 2017;35:441‐468. 10.1146/annurev-immunol-051116-052358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Hickman S, Izzy S, Sen P, Morsett L, El Khoury J. Microglia in neurodegeneration. Nat Neurosci. 2018;21(10):1359‐1369. 10.1038/s41593-018-0242-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Favuzzi E, Huang S, Saldi GA, et al. GABA‐receptive microglia selectively sculpt developing inhibitory circuits. Cell. 2021;184(15):4048‐4063. 10.1016/j.cell.2021.06.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Lee JH, Yang DS, Goulbourne CN, et al. Faulty autolysosome acidification in Alzheimer's disease mouse models induces autophagic build‐up of Aβ in neurons, yielding senile plaques. Nat Neurosci. 2022;25(6):688‐701. 10.1038/s41593-022-01084-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Tejera D, Heneka MT. Microglia in neurodegenerative disorders. Methods Mol Biol. 2019;2034:57‐67. 10.1007/978-1-4939-9658-2_5 [DOI] [PubMed] [Google Scholar]
  • 66. Fu R, Shen Q, Xu P, Luo JJ, Tang Y. Phagocytosis of microglia in the central nervous system diseases. Mol Neurobiol. 2014;49(3):1422‐1434. 10.1007/s12035-013-8620-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122(3):787‐795. 10.1172/jci59643 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Tang Y, Le W. Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol Neurobiol. 2016;53(2):1181‐1194. 10.1007/s12035-014-9070-5 [DOI] [PubMed] [Google Scholar]
  • 69. Kalafatakis I, Karagogeos D. Oligodendrocytes and microglia: key players in myelin development, damage and repair. Biomolecules. 2021;11(7):1058. 10.3390/biom11071058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Kwon M‐S. Advanced therapeutic strategies targeting microglia: beyond neuroinflammation. Arch Pharmacal Res. 2022;45(9):618‐630. 10.1007/s12272-022-01406-1 [DOI] [PubMed] [Google Scholar]
  • 71. Irfan M, Evonuk KS, DeSilva TM. Microglia phagocytose oligodendrocyte progenitor cells and synapses during early postnatal development: implications for white versus gray matter maturation. FEBS J. 2022;289(8):2110‐2127. 10.1111/febs.16190 [DOI] [PubMed] [Google Scholar]
  • 72. Green KN, Crapser JD, Hohsfield LA. To kill a microglia: a case for CSF1R inhibitors. Trends Immunol. 2020;41(9):771‐784. 10.1016/j.it.2020.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Willis EF, MacDonald KPA, Nguyen QH, et al. Repopulating microglia promote brain repair in an IL‐6‐dependent manner. Cell. 2020;180(5):833‐846. 10.1016/j.cell.2020.02.013 [DOI] [PubMed] [Google Scholar]
  • 74. Li Y, He X, Kawaguchi R, et al. Microglia‐organized scar‐free spinal cord repair in neonatal mice. Nature. 2020;587(7835):613‐618. 10.1038/s41586-020-2795-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Brennan FH, Li Y, Wang C, et al. Microglia coordinate cellular interactions during spinal cord repair in mice. Nat Commun. 2022;13(1):4096. 10.1038/s41467-022-31797-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Xu Z, Rao Y, Huang Y, et al. Efficient strategies for microglia replacement in the central nervous system. Cell Rep. 2020;32(6):108041. 10.1016/j.celrep.2020.108041 [DOI] [PubMed] [Google Scholar]
  • 77. Hong DS, Deeg HJ. Hemopoietic stem cells: sources and applications. Med Oncol. 1994;11(2):63‐68. [DOI] [PubMed] [Google Scholar]
  • 78. Ogawa M. Hemopoietic stem cells: stochastic differentiation and humoral control of proliferation. Environ Health Perspect. 1989;80:199‐207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Fliedner TM, Fliedner TM. The role of blood stem cells in hematopoietic cell renewal. Stem Cells. 1998;16(suppl 1):13‐29. [DOI] [PubMed] [Google Scholar]
  • 80. Höfer T, Rodewald H‐R. Differentiation‐based model of hematopoietic stem cell functions and lineage pathways. Blood. 2018;132(11):1106‐1113. 10.1182/blood-2018-03-791517 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Ginhoux F, Greter M, Leboeuf M, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010;330(6005):841‐845. 10.1126/science.1194637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Shibuya Y, Kumar KK, Mader MM‐D, et al. Treatment of a genetic brain disease by CNS‐wide microglia replacement. Sci Transl Med. 2022;14(636):eabl9945. 10.1126/scitranslmed.abl9945 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Larochelle A, Bellavance M‐A, Michaud J‐P, Rivest S. Bone marrow‐derived macrophages and the CNS: an update on the use of experimental chimeric mouse models and bone marrow transplantation in neurological disorders. Biochim Biophys Acta. 2016;1862(3):310‐322. 10.1016/j.bbadis.2015.09.017 [DOI] [PubMed] [Google Scholar]
  • 84. Mishra P, Silva A, Sharma J, et al. Rescue of Alzheimer's disease phenotype in a mouse model by transplantation of wild‐type hematopoietic stem and progenitor cells. Cell Rep. 2023;42(8):112956. 10.1016/j.celrep.2023.112956 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Atkins H. Stem cell transplantation to treat multiple sclerosis. JAMA. 2019;321(2):153‐155. 10.1001/jama.2018.20777 [DOI] [PubMed] [Google Scholar]
  • 86. Savitz SI, Rosenbaum DM, Dinsmore JH, Wechsler LR, Caplan LR. Cell transplantation for stroke. Ann Neurol. 2002;52(3):266‐275. [DOI] [PubMed] [Google Scholar]
  • 87. SanMartin A, Borlongan CV. Article commentary: cell transplantation: toward cell therapy. Cell Transplant. 2006;15(7):665‐673. [DOI] [PubMed] [Google Scholar]
  • 88. Ferreira LMR, Muller YD, Bluestone JA, Tang Q. Next‐generation regulatory T cell therapy. Nat Rev Drug Discov. 2019;18(10):749‐769. 10.1038/s41573-019-0041-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Cai S, Chandraker A. Cell therapy in solid organ transplantation. Curr Gene Ther. 2019;19(2):71‐80. 10.2174/1566523219666190603103840 [DOI] [PubMed] [Google Scholar]
  • 90. Hu M, Hawthorne WJ, Yi S, O'Connell PJ. Cellular immune responses in islet xenograft rejection. Front Immunol. 2022;13:893985. 10.3389/fimmu.2022.893985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Esquirol A, Cadenas IG, Novelli S, et al. Outcome improvement over time in reduced intensity conditioning hematopoietic transplantation: a 20‐year experience. Ann Hematol. 2023;103:321‐334. 10.1007/s00277-023-05530-w [DOI] [PubMed] [Google Scholar]
  • 92. Hooks MA. Tacrolimus, a new immunosuppressant—a review of the literature. Ann Pharmacother. 1994;28(4):501‐511. [DOI] [PubMed] [Google Scholar]
  • 93. Zhao Y, Shen X‐F, Cao K, et al. Dexamethasone‐induced myeloid‐derived suppressor cells prolong allo cardiac graft survival through iNOS‐ and glucocorticoid receptor‐dependent mechanism. Front Immunol. 2018;9:282. 10.3389/fimmu.2018.00282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Roberts MB, Fishman JA. Immunosuppressive agents and infectious risk in transplantation: managing the “net state of immunosuppression”. Clin Infect Dis. 2021;73(7):e1302‐e1317. 10.1093/cid/ciaa1189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Mecadon K. Managing side effects of immunosuppressants. R I Med J. 2021;104(1):31‐33. [PubMed] [Google Scholar]
  • 96. Wojciechowski D, Wiseman A. Long‐term immunosuppression management: opportunities and uncertainties. Clin J Am Soc Nephrol. 2021;16(8):1264‐1271. 10.2215/CJN.15040920 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Park TY, Jeon J, Lee N, et al. Co‐transplantation of autologous Treg cells in a cell therapy for Parkinson's disease. Nature. 2023;619:606‐615. 10.1038/s41586-023-06300-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Morizane A, Kikuchi T, Hayashi T, et al. MHC matching improves engraftment of iPSC‐derived neurons in non‐human primates. Nat Commun. 2017;8:385. 10.1038/s41467-017-00926-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Brett BL, Gardner RC, Godbout J, Dams‐O'Connor K, Keene CD. Traumatic brain injury and risk of neurodegenerative disorder. Biol Psychiatry. 2022;91(5):498‐507. 10.1016/j.biopsych.2021.05.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Théry C, Amigorena S. The cell biology of antigen presentation in dendritic cells. Curr Opin Immunol. 2001;13(1):45‐51. [DOI] [PubMed] [Google Scholar]
  • 101. Austyn JM. Dendritic cells. Curr Opin Hematol. 1998;5(1):3‐15. [DOI] [PubMed] [Google Scholar]
  • 102. Saas P, Tiberghien P. Dendritic cells: to where do they lead? Transplantation. 2002;73(suppl 1):S12‐S15. [DOI] [PubMed] [Google Scholar]
  • 103. Hilligan KL, Ronchese F. Antigen presentation by dendritic cells and their instruction of CD4+ T helper cell responses. Cell Mol Immunol. 2020;17(6):587‐599. 10.1038/s41423-020-0465-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Kotsias F, Cebrian I, Alloatti A. Antigen processing and presentation. Int Rev Cell Mol Biol. 2019;348:69‐121. 10.1016/bs.ircmb.2019.07.005 [DOI] [PubMed] [Google Scholar]
  • 105. Mundt S, Greter M, Becher B. The CNS mononuclear phagocyte system in health and disease. Neuron. 2022;110(21):3497‐3512. 10.1016/j.neuron.2022.10.005 [DOI] [PubMed] [Google Scholar]
  • 106. Subbarayan MS, Hudson C, Moss LD, Nash KR, Bickford PC. T cell infiltration and upregulation of MHCII in microglia leads to accelerated neuronal loss in an α‐synuclein rat model of Parkinson's disease. J Neuroinflammation. 2020;17(1):242. 10.1186/s12974-020-01911-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Aron Badin R, Bugi A, Williams S, et al. MHC matching fails to prevent long‐term rejection of iPSC‐derived neurons in non‐human primates. Nat Commun. 2019;10:4357. 10.1038/s41467-019-12324-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Schonhoff AM, Figge DA, Williams GP, et al. Border‐associated macrophages mediate the neuroinflammatory response in an alpha‐synuclein model of Parkinson disease. Nat Commun. 2023;14(1):3754. 10.1038/s41467-023-39060-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Baumans V. Use of animals in experimental research: an ethical dilemma? Gene Ther. 2004;11(suppl 1):S64‐S66. [DOI] [PubMed] [Google Scholar]
  • 110. Kang M, Long T, Chang C, et al. A review of the ethical use of animals in functional experimental research in China based on the “Four R” principles of Reduction, Replacement, Refinement, and Responsibility. Med Sci Monit. 2022;29:e938807. 10.12659/MSM.938807 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. De Niz M, Heussler VT. Rodent malaria models: insights into human disease and parasite biology. Curr Opin Microbiol. 2018;46:93‐101. 10.1016/j.mib.2018.09.003 [DOI] [PubMed] [Google Scholar]
  • 112. Habert R, Muczynski V, Grisin T, et al. Concerns about the widespread use of rodent models for human risk assessments of endocrine disruptors. Reproduction. 2014;147(4):R119‐R129. 10.1530/REP-13-0497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Ito R, Lee ACH. The role of the hippocampus in approach‐avoidance conflict decision‐making: evidence from rodent and human studies. Behav Brain Res. 2016;313:345‐357. 10.1016/j.bbr.2016.07.039 [DOI] [PubMed] [Google Scholar]
  • 114. Beauchamp A, Yee Y, Darwin BC, Raznahan A, Mars RB, Lerch JP. Whole‐brain comparison of rodent and human brains using spatial transcriptomics. eLife. 2022;11:e79418. 10.7554/eLife.79418 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Charvet CJ, Ofori K, Baucum C, et al. Tracing modification to cortical circuits in human and nonhuman primates from high‐resolution tractography, transcription, and temporal dimensions. J Neurosci. 2022;42(18):3749‐3767. 10.1523/JNEUROSCI.1506-21.2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Eichert N, Robinson EC, Bryant KL, et al. Cross‐species cortical alignment identifies different types of anatomical reorganization in the primate temporal lobe. eLife. 2020;9:e53232. 10.7554/eLife.53232 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Kret ME, Prochazkova E, Sterck EHM, Clay Z. Emotional expressions in human and non‐human great apes. Neurosci Biobehav Rev. 2020;115:378‐395. 10.1016/j.neubiorev.2020.01.027 [DOI] [PubMed] [Google Scholar]
  • 118. Rilling JK. Comparative primate neurobiology and the evolution of brain language systems. Curr Opin Neurobiol. 2014;28:10‐14. 10.1016/j.conb.2014.04.002 [DOI] [PubMed] [Google Scholar]
  • 119. Roth G, Dicke U. Origin and evolution of human cognition. Prog Brain Res. 2019;250:285‐316. 10.1016/bs.pbr.2019.02.004 [DOI] [PubMed] [Google Scholar]
  • 120. Marchetto MCN, Narvaiza I, Denli AM, et al. Differential L1 regulation in pluripotent stem cells of humans and apes. Nature. 2013;503(7477):525‐529. 10.1038/nature12686 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Kanton S, Boyle MJ, He Z, et al. Organoid single‐cell genomic Atlas uncovers human‐specific features of brain development. Nature. 2019;574(7778):418‐422. 10.1038/s41586-019-1654-9 [DOI] [PubMed] [Google Scholar]
  • 122. Clevers H. Modeling development and disease with organoids. Cell. 2016;165(7):1586‐1597. 10.1016/j.cell.2016.05.082 [DOI] [PubMed] [Google Scholar]
  • 123. Corrò C, Novellasdemunt L, Li VSW. A brief history of organoids. Am J Physiol Cell Physiol. 2020;319(1):C151‐C165. 10.1152/ajpcell.00120.2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Lancaster MA, Renner M, Martin CA, et al. Cerebral organoids model human brain development and microcephaly. Nature. 2013;501(7467):373‐379. 10.1038/nature12517 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Thomson JA, Itskovitz‐Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145‐1147. 10.1126/science.282.5391.1145 [DOI] [PubMed] [Google Scholar]
  • 126. Moscona A. Rotation‐mediated histogenetic aggregation of dissociated cells. Exp Cell Res. 1961;22:455‐475. 10.1016/0014-4827(61)90122-7 [DOI] [PubMed] [Google Scholar]
  • 127. Zhang SC, Wernig M, Duncan ID, Brüstle O, Thomson JA. In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nat Biotechnol. 2001;19(12):1129‐1133. 10.1038/nbt1201-1129 [DOI] [PubMed] [Google Scholar]
  • 128. Nakano T, Ando S, Takata N, et al. Self‐formation of optic cups and storable stratified neural retina from human ESCs. Cell Stem Cell. 2012;10(6):771‐785. 10.1016/j.stem.2012.05.009 [DOI] [PubMed] [Google Scholar]
  • 129. Dorszewska J, Prendecki M, Oczkowska A, Dezor M, Kozubski W. Molecular basis of familial and sporadic Alzheimer's disease. Curr Alzheimer Res. 2016;13(9):952‐963. 10.2174/1567205013666160314150501 [DOI] [PubMed] [Google Scholar]
  • 130. Nagatsu T. Catecholamines and Parkinson's disease: tyrosine hydroxylase (TH) over tetrahydrobiopterin (BH4) and GTP cyclohydrolase I (GCH1) to cytokines, neuromelanin, and gene therapy: a historical overview. J Neural Transm. Published online August 28, 2023. 10.1007/s00702-023-02673-y [DOI] [PubMed]
  • 131. Dong X, Xu S‐B, Chen X, et al. Human cerebral organoids establish subcortical projections in the mouse brain after transplantation. Mol Psychiatry. 2021;26(7):2964‐2976. 10.1038/s41380-020-00910-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Revah O, Gore F, Kelley KW, et al. Maturation and circuit integration of transplanted human cortical organoids. Nature. 2022;610(7931):319‐326. 10.1038/s41586-022-05277-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Cao S‐Y, Yang D, Huang Z‐Q, et al. Cerebral organoids transplantation repairs infarcted cortex and restores impaired function after stroke. NPJ Regen Med. 2023;8(1):27. 10.1038/s41536-023-00301-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Cao SY, Yang D, Huang ZQ, et al. Cerebral organoids transplantation repairs infarcted cortex and restores impaired function after stroke. NPJ Regen Med. 2023;8(1):27. 10.1038/s41536-023-00301-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Li Y, Huang J, Wang J, et al. Human umbilical cord‐derived mesenchymal stem cell transplantation supplemented with curcumin improves the outcomes of ischemic stroke via AKT/GSK‐3β/β‐TrCP/Nrf2 axis. J Neuroinflammation. 2023;20(1):49. 10.1186/s12974-023-02738-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Jgamadze D, Lim JT, Zhang Z, et al. Structural and functional integration of human forebrain organoids with the injured adult rat visual system. Cell Stem Cell. 2023;30(2):137‐152. 10.1016/j.stem.2023.01.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Wilson MN, Thunemann M, Liu X, et al. Multimodal monitoring of human cortical organoids implanted in mice reveal functional connection with visual cortex. Nat Commun. 2022;13(1):7945. 10.1038/s41467-022-35536-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Mansour AA, Gonçalves JT, Bloyd CW, et al. An in vivo model of functional and vascularized human brain organoids. Nat Biotechnol. 2018;36(5):432‐441. 10.1038/nbt.4127 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Heemels M‐T. Neurodegenerative diseases. Nature. 2016;539(7628):179. 10.1038/539179a [DOI] [PubMed] [Google Scholar]
  • 140. Das M, Mayilsamy K, Mohapatra SS, Mohapatra S. Mesenchymal stem cell therapy for the treatment of traumatic brain injury: progress and prospects. Rev Neurosci. 2019;30(8):839‐855. 10.1515/revneuro-2019-0002 [DOI] [PubMed] [Google Scholar]
  • 141. Nakamura R, Patel BA, Kim S, et al. Conditional survival and standardized mortality ratios of patients with severe aplastic anemia surviving at least one year after hematopoietic cell transplantation or immunosuppressive therapy. Haematologica. 2023;108(12):3298‐3307. 10.3324/haematol.2023.282781 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Wu J, Zheng Z, Chong Y, et al. Immune responsive release of tacrolimus to overcome organ transplant rejection. Adv Mater. 2018;30(45):e1805018. 10.1002/adma.201805018 [DOI] [PubMed] [Google Scholar]
  • 143. de Jongh D, Massey EK, Berishvili E, et al. Organoids: a systematic review of ethical issues. Stem Cell Res Ther. 2022;13(1):337. 10.1186/s13287-022-02950-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Trujillo CA, Muotri AR. Brain organoids and the study of neurodevelopment. Trends Mol Med. 2018;24(12):982‐990. 10.1016/j.molmed.2018.09.005 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from Ibrain are provided here courtesy of Affiliated Hospital of Zunyi Medical University (AHZMU) and Wiley-VCH GmbH

RESOURCES