Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1934 2022-11-14 14:40:55 |
2 format corrected. Meta information modification 1934 2022-11-15 03:05:35 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zayed, M.A.;  Sultan, S.;  Alsaab, H.O.;  Yousof, S.M.;  Alrefaei, G.I.;  Alsubhi, N.H.;  Alkarim, S.;  Ghamdi, K.S.A.;  Bagabir, S.A.;  Jana, A.; et al. Stem-Cell-Based Therapy in Alzheimer’s Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/34487 (accessed on 03 July 2024).
Zayed MA,  Sultan S,  Alsaab HO,  Yousof SM,  Alrefaei GI,  Alsubhi NH, et al. Stem-Cell-Based Therapy in Alzheimer’s Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/34487. Accessed July 03, 2024.
Zayed, Mohamed A., Samar Sultan, Hashem O. Alsaab, Shimaa Mohammad Yousof, Ghadeer I. Alrefaei, Nouf H. Alsubhi, Saleh Alkarim, Kholoud S. Al Ghamdi, Sali Abubaker Bagabir, Ankit Jana, et al. "Stem-Cell-Based Therapy in Alzheimer’s Disease" Encyclopedia, https://encyclopedia.pub/entry/34487 (accessed July 03, 2024).
Zayed, M.A.,  Sultan, S.,  Alsaab, H.O.,  Yousof, S.M.,  Alrefaei, G.I.,  Alsubhi, N.H.,  Alkarim, S.,  Ghamdi, K.S.A.,  Bagabir, S.A.,  Jana, A.,  Alghamdi, B.S.,  Atta, H.M., & Ashraf, G.M. (2022, November 14). Stem-Cell-Based Therapy in Alzheimer’s Disease. In Encyclopedia. https://encyclopedia.pub/entry/34487
Zayed, Mohamed A., et al. "Stem-Cell-Based Therapy in Alzheimer’s Disease." Encyclopedia. Web. 14 November, 2022.
Stem-Cell-Based Therapy in Alzheimer’s Disease
Edit

Stem cells are a versatile source for cell therapy. Their use is particularly significant for the treatment of neurological disorders for which no definitive conventional medical treatment is available. Neurological disorders are of diverse etiology and pathogenesis. Alzheimer’s disease (AD) is caused by abnormal protein deposits, leading to progressive dementia.

stem cells therapy neurodegenerative diseases Parkinson’s disease

1. Introduction

In the 21st century, stem cells have gained tremendous importance in the fields of medical research and therapy. Stem cells are recognized as body cells that have unique characteristics, including the ability of self-renewal and differentiation into several type of body cells [1]. They can remain undifferentiated (totipotent) and are capable of differentiating into several mature cells [2]. Stem cells can be categorized depending on their sources: embryonic stem cells, fetal stem cells, adult stem cells and induced pluripotent stem cells [1].
In the medical field, stem cells have been authorized for use in bone marrow transplantation for the treatment of hematological malignancies and some inherited metabolic diseases. Recently, successful stem cell transplantation was reported to cure human immunodeficiency virus (HIV) infection [3]. Several experimental studies and/or clinical trials studied the use of different kinds of stem cells for the treatment of neurological disorders particularly the disorders lacking a definitive medical treatment. These include Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s Disease (HD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), temporal lobe epilepsy (TLE), neuropathic pain (NP), and brain ischemic stroke (BIS) [4][5].
The significance of stem cells is derived from their reported ability to replenish damaged cells and tissues as well as their anti-inflammatory and immune-modulatory properties. Stem cells can differentiate and replenish cells that have been weakened or destroyed, promoting neural tissue growth and development. Most of the neurological disorders are characterized by the widespread neuronal death and the extremely low regenerative potential of the brain. The treatment needs materials or cells that can cross the blood brain barrier (BBB). All these factors contribute to making stem cell therapy a viable option for treating chronic intractable neurologic diseases [6][7]
By combining additional drugs, the results of stem cell therapy may be improved [8]. Stem cell treatment, for example, in combination with erythropoietin, had synergetic effects on rat neurogenesis. To overcome the limitations of stem cell migration and inclusion in functional networks [9][10][11], nanoparticle distribution systems are investigated. Since they cross the BBB and enter the target brain areas without affecting the surroundings, these nanoparticles are beneficial for drug and cell systems. Another choice for delivering and preserving stem cells in the transplant site is hydrophilic polymer encapsulation, thereby providing mechanical assistance in supply processes and increasing the proliferation and differentiation in hydrogels [12]. In recent research, gene therapy and neural development factors have also been used to extend the retention of AD and PD transplanted stem cells [13].
Many recent trials have shown the value of peripheral stem cell treatments for acute stroke survivors, enabling minimally invasive cell therapy to become a practical alternative option. Additionally, there is consideration of the use of mesenchymal stem cells (MSCs) for supplying bioactive factors, such as brain-derived neurotrophic factors (BDNF), in treating neurologic disorders, such as HD [14][15][16].

2. Selection of Transplant Recipients

Cell transplantation requires several sets of issues that must be considered in preclinical and clinical trials [17]. The first is whether the disease causes brain cell death or initiates a transition in cell interactions. The next is the probability of systemic transplantation, which happens only if the blood–brain barrier (BBB) is known to be open. Another aspect is whether pathology causes an inflammatory response in relation to the condition itself. In this case, the transplanted cells can play not only an alternative role, but also an anti-inflammatory role. All critical issues must be considered for nearly all cells used in clinical trials [17].
Given the above, it is not easy to choose the cells to be transplanted. Many various stem cell types play a possible therapeutic role in the treatment of neurological conditions. Cells must play either a substitutional or trophic role. There was a great focus on the substitution feature in earlier years of stem cell transplantation in neurological disorders. Many experimental studies in animal models showed that transplanted cells in the nerve tissue did not produce a physiological response [18][19]. If the neural dysfunction is still significant and the surgical approach takes place, the tissue’s reconstitution and hence the rebuilding of the injured neural pathways are very complex. Spinal cord damage is associated with the loss of motor neurons with long axons covered by the myelin sheath. In such circumstances, the transplanted cells must replenish neurons and glia; however, to exert their therapeutic activity, they must be capable of expanding their processes in the right direction. It is unlikely that this challenge will be completed with the current expertise, but a potential approach for transplant research in sophisticated tissues may be the fusion of transplant therapy and bioengineering (scaffold construction) [12].
No biomarkers or successful medicines were able to slow down disease development, despite the billions in dollars of clinical trials and considerable advances in studying neurodegenerative mechanisms. This makes stem cell therapy for the treatment of neurodegenerative diseases a beneficial approach to be tested [20]. The first aim of stem cell therapy involves determining distinct neuronal subtypes and the recapitulation of a network of neural diseases similar to those lost. The development of environmental reinforcement in support of host neurons by the production of neurotrophic and scavenging toxic factors and the construction of an auxiliary neural network across the affected areas is another approach to the treatment of neurodegenerative disorders [21]. Another strategic approach is the synthesis of neuroprotective growth factors in the sites of diseases (e.g., glial-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor (BDNF), the insulin-like factor 1 growth factor (IGF-1), and VEGF).
One of the biggest concerns is immune rejection of transplanted fetal tissue or cells, which can trigger serious host responses [22]. Although the brain is considered immuno-privileged, few human leucocyte antigen cells remain matched to the haplotype, requiring immunosuppression in recipients to prevent cell-induced immune refusal. While there are a few exceptions, new technologies are necessary to increase donors’ and recipients’ compatibility and avoid further immune rejections [23][24].

3. Stem Cells in Alzheimer’s Disease (AD)

Alzheimer’s disease is a widespread chronic, pathologically marked neurodegenerative condition with ß-amyloid plaques and neurofibrillary tangles. Current alternatives to medication only relieve the symptoms without treating the illness, which is a significant problem that impacts the quality of life of patients and their care providers. Stem cell therapy may offer new opportunities for AD patients’ care. More and more research has shown that neural stem cells(NSCs) developed from embryonic stem cells (ESCs) were efficient as a treatment approach in AD models, showing changes both in vitro and in vivo [25][26]. Stem cells have the potential to differentiate from the brain extracellular matrix into neural cells, and they may restore neuroplasticity and neurogenesis via neurotrophic factors [27].
The stem cell approach to treating AD was first examined in animal models [28]. Neural stem cells from neonatal rat brains were used to establish new cholinergic neurons and increased learning and memory in rats with AD [29]. Neuron-like embryonic stem cells were used to restore AD-damaged rat brains [30]. Lately, the most used cells in Alzheimer’s disease research were embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), brain-derived neural stem cells (NSCs), and induced pluripotent stem cells (iPSCs) [31].
The basal forebrain cholinergic neurons (BFCNs) are critically implicated in memory and learning disorders, such as AD [32]. The ESCs possess the pluripotency potential, which is double-bladed. Although that pluripotency is a great advantage for ESCs, it is a considerable disadvantage, as it may lead to the differentiation of the ESCs into several directions, which ultimately can lead to the formation of teratomas and tumors. Moreover, there is a tendency for eliciting disturbed immune reactions and rejection on transplanting ESCs [33][34]. Therefore, although the ESCs showed promising results in rat models of AD and improved memory performance, it has limited clinical applications. Two types of ESCs have been used in AD research: mouse ESCs (mESCs) and human ESCs (hESCs). Both produced the differentiation into BFCNs when transplanted in mice models with AD.
The immune rejection that occurs using allogeneic hESCs can be mitigated by transplanting iPSCs. Therefore, stimulating the differentiation of autologous hiPSCs could be promising in diminishing the chances of such immune rejection. Nevertheless, there are many concerns regarding the safety of using the iPSCs including the potential risk of oncogenesis and teratoma formation, the safety of the long-term use, the reprogramming efficiency, and immunogenic liability. The partial reprogramming and the unstable genes might elicit an immunological reaction with iPSCs. There is a need to develop new methods and protocols to avoid the expression of the tumorigenic genes when using iPSCs [34][35]. The iPSCs can differentiate into different cell types, including neurons. In AD research, iPSCs can be used, for example, to investigate the inflammatory reaction, to induce macrophages that can express a protease that degrades beta-amyloid called neprilysin and to reprogram the fibroblast and hence identify the phenotype of the AD [31][36][37].
Despite the ethical issues, the most widely used type of stem cells utilized in AD research is MSCs obtained from umbilical cord blood. This is attributed to the feasibility of obtaining umbilical cord blood after delivery [31][38]. Previous reports have shown that MSCs can improve the deficits in memory and learning in AD murine models. Boutajangout et al. reported that human umbilical cord mesenchymal stem cell (HUC-MSCs) xenografts improved cognitive decline and reduced the Amyloid burden in a mouse model of Alzheimer’s disease [39].
Many mechanisms have been suggested to be involved in this process, including decreased beta-amyloid plaques, a dramatic decrease in β-secretase 1 (BACE-1) levels, reduced hyperphosphorylation of tau, and the reversal of the inflammatory process in the microglia as well as the enhancement of anti-inflammatory cytokines [40]. Additionally, the immunomodulation and anti-inflammatory effects of MSCs have been reported to occur via enhancing the neuroprotection and depressing the proinflammatory cytokines. Moreover, bone marrow MSCs have been found to stimulate the formation of extracellular vesicles and microvesicles. These vesicles, in turn, target the amyloid-beta [41][42]. Additionally, the evolved plasticity and neurotrophic decline, decreased tau phosphorylation, and neuroinflammation, and tau down-regulation are promising targets for stem cells. Results showed the transgenic mice survived without any harmful effects and showed increased memory. This was confirmed in another study, where synaptogenesis increased the mental capacity in mice [43][44]. The successful preliminary animal studies showed positive findings. Researchers grafted human umbilical mesenchymal stem cells obtained from donor cords, into AD mice. An anti-inflammatory and immune-modulatory reaction was simultaneously induced in the mice by this research, and the presence of M2-like microglia enhanced synapsin and raised A β levels in the brain, thereby decreasing amyloid accumulation [45]. This led to a clinical trial in 2015 using human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) on nine patients with mild-to-to-moderate AD. The hUCB-MSCs were stereotactically inserted into the hippocampus. The method of administration of the stem cells was stable and feasible, with no consequences. Better studies with larger sample size and placebo monitoring are needed to advance the hypothesis. The administration of stem cell therapy was safe but needs to be further checked for its therapeutic efficacy on AD pathogenesis. [46]. The outcome of some studies on Alzheimer’s patients is still unknown such as (NTC01547689, NTC02672306, NTC02054208, and NTC02600130 from Clinicaltrials.gov). Nevertheless, they are all experiments constrained by the variation of neurons affected by AD.
The biotechnology company Nature Cell has begun a new phase II clinical trial using a stem cell medicine for AD (AstroStem) consisting of autologous adipose tissue stem cells administered intravenously into 60 AD patients (200 million cells/injection) (NCT03117738).

References

  1. Hui, H.; Tang, Y.; Hu, M.; Zhao, X. Stem Cells: General Features and Characteristics; IntechOpen: London, UK, 2011.
  2. Mitalipov, S.; Wolf, D. Totipotency, pluripotency and nuclear reprogramming. Adv. Biochem Eng. Biotechnol 2009, 114, 185–199.
  3. Gupta, R.K.; Peppa, D.; Hill, A.L.; Gálvez, C.; Salgado, M.; Pace, M.; McCoy, L.E.; Griffith, S.A.; Thornhill, J.; Alrubayyi, A.; et al. Evidence for HIV-1 cure after CCR5Δ32/Δ32 allogeneic haemopoietic stem-cell transplantation 30 months post analytical treatment interruption: A case report. Lancet HIV 2020, 7, e340–e347.
  4. Biehl, J.K.; Russell, B. Introduction to stem cell therapy. J. Cardiovasc. Nurs. 2009, 24, 98–103.
  5. Conese, M.; Piro, D.; Carbone, A.; Castellani, S.; Di Gioia, S. Hematopoietic and Mesenchymal Stem Cells for the Treatment of Chronic Respiratory Diseases: Role of Plasticity and Heterogeneity. Sci. World J. 2014, 2014, 859817.
  6. Hamblin, M.R. Photobiomodulation for Alzheimer’s Disease: Has the Light Dawned? Photonics 2019, 6, 77.
  7. Sivandzade, F.; Cucullo, L. Regenerative Stem Cell Therapy for Neurodegenerative Diseases: An Overview. Int. J. Mol. Sci. 2021, 22, 2153.
  8. Ritfeld, G.J.; Roos, R.A.; Oudega, M. Stem cells for central nervous system repair and rehabilitation. PMR 2011, 3, S117–S122.
  9. Carballo-Molina, O.A.; Sánchez-Navarro, A.; López-Ornelas, A.; Lara-Rodarte, R.; Salazar, P.; Campos-Romo, A.; Ramos-Mejía, V.; Velasco, I. Semaphorin 3C released from a biocompatible hydrogel guides and promotes axonal growth of rodent and human dopaminergic neurons. Tissue Eng. Part. A 2016, 22, 850–861.
  10. Garcia-Bennett, A.E.; König, N.; Abrahamsson, N.; Kozhevnikova, M.; Zhou, C.; Trolle, C.; Pankratova, S.; Berezin, V.; Kozlova, E.N. In vitro generation of motor neuron precursors from mouse embryonic stem cells using mesoporous nanoparticles. Nanomedicine 2014, 9, 2457–2466.
  11. Kuo, Y.-C.; Rajesh, R. Nerve growth factor-loaded heparinized cationic solid lipid nanoparticles for regulating membrane charge of induced pluripotent stem cells during differentiation. Mater. Sci. Eng. C 2017, 77, 680–689.
  12. Zhu, W.; George, J.K.; Sorger, V.J.; Zhang, L.G. 3D printing scaffold coupled with low level light therapy for neural tissue regeneration. Biofabrication 2017, 9, 025002.
  13. Li, Q.; Wang, Z.; Xing, H.; Wang, Y.; Guo, Y. Exosomes derived from miR-188-3p-modified adipose-derived mesenchymal stem cells protect Parkinson’s disease. Mol. Ther. -Nucleic Acids 2021, 23, 1334–1344.
  14. Deng, P.; Anderson, J.D.; Yu, A.S.; Annett, G.; Fink, K.D.; Nolta, J.A. Engineered BDNF producing cells as a potential treatment for neurologic disease. Expert Opin. Biol. Ther. 2016, 16, 1025–1033.
  15. Fink, K.D.; Deng, P.; Gutierrez, J.; Anderson, J.S.; Torrest, A.; Komarla, A.; Kalomoiris, S.; Cary, W.; Anderson, J.D.; Gruenloh, W. Allele-specific reduction of the mutant huntingtin allele using transcription activator-like effectors in human huntington’s disease fibroblasts. Cell Transplant. 2016, 25, 677–686.
  16. Nolta, J.A. “Next-Generation” Mesenchymal Stem or Stromal Cells for the In Vivo Delivery of Bioactive Factors: Progressing Toward the Clinic. Transfusion 2016, 56, 15S–17S.
  17. Adami, R.; Scesa, G.; Bottai, D. Stem cell transplantation in neurological diseases: Improving effectiveness in animal models. Front. Cell Dev. Biol. 2014, 2, 17.
  18. Cusimano, M.; Biziato, D.; Brambilla, E.; Donegà, M.; Alfaro-Cervello, C.; Snider, S.; Salani, G.; Pucci, F.; Comi, G.; Garcia-Verdugo, J.M.; et al. Transplanted neural stem/precursor cells instruct phagocytes and reduce secondary tissue damage in the injured spinal cord. Brain A J. Neurol. 2012, 135, 447–460.
  19. Nizzardo, M.; Simone, C.; Rizzo, F.; Ruggieri, M.; Salani, S.; Riboldi, G.; Faravelli, I.; Zanetta, C.; Bresolin, N.; Comi, G.P. Minimally invasive transplantation of iPSC-derived ALDHhiSSCloVLA4+ neural stem cells effectively improves the phenotype of an amyotrophic lateral sclerosis model. Hum. Mol. Genet. 2014, 23, 342–354.
  20. Karussis, D.; Petrou, P.; Kassis, I. Clinical experience with stem cells and other cell therapies in neurological diseases. J. Neurol. Sci. 2013, 324, 1–9.
  21. Lunn, J.S.; Sakowski, S.A.; Hur, J.; Feldman, E.L. Stem cell technology for neurodegenerative diseases. Ann. Neurol. 2011, 70, 353–361.
  22. Zhao, T.; Zhang, Z.-n.; Westenskow, P.D.; Todorova, D.; Hu, Z.; Lin, T.; Rong, Z.; Kim, J.; He, J.; Wang, M. Humanized mice reveal differential immunogenicity of cells derived from autologous induced pluripotent stem cells. Cell Stem Cell 2015, 17, 353–359.
  23. Chen, Y.; Pan, C.; Xuan, A.; Xu, L.; Bao, G.; Liu, F.; Fang, J.; Long, D. Treatment efficacy of NGF nanoparticles combining neural stem cell transplantation on Alzheimer’s disease model rats. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2015, 21, 3608.
  24. Hallett, P.J.; Cooper, O.; Sadi, D.; Robertson, H.; Mendez, I.; Isacson, O. Long-term health of dopaminergic neuron transplants in Parkinson’s disease patients. Cell Rep. 2014, 7, 1755–1761.
  25. Wang, F.; Jia, Y.; Liu, J.; Zhai, J.; Cao, N.; Yue, W.; He, H.; Pei, X. Dental pulp stem cells promote regeneration of damaged neuron cells on the cellular model of Alzheimer’s disease. Cell Biol. Int. 2017, 41, 639–650.
  26. Zhao, H. Embryonic neural stem cell transplantation for Alzheimer’s disease. Chin. J. Tissue Eng. Res. 2016, 20, 4805–4810.
  27. Enciu, A.M.; Nicolescu, M.I.; Manole, C.G.; Mureşanu, D.F.; Popescu, L.M.; Popescu, B.O. Neuroregeneration in neurodegenerative disorders. BMC Neurol. 2011, 11, 1–7.
  28. Kang, J.M.; Yeon, B.K.; Cho, S.J.; Suh, Y.H. Stem Cell Therapy for Alzheimer’s Disease: A Review of Recent Clinical Trials. J. Alzheimer’s Dis. JAD 2016, 54, 879–889.
  29. Xuan, A.; Luo, M.; Ji, W.; Long, D. Effects of engrafted neural stem cells in Alzheimer’s disease rats. Neurosci. Lett. 2009, 450, 167–171.
  30. Hoveizi, E.; Mohammadi, T.; Moazedi, A.A.; Zamani, N.; Eskandary, A. Transplanted neural-like cells improve memory and Alzheimer-like pathology in a rat model. Cytotherapy 2018, 20, 964–973.
  31. Liu, M.; Li, K.; Wang, Y.; Zhao, G.; Jiang, J. Stem cells in the treatment of neuropathic pain: Research progress of mechanism. Stem Cells Int 2020, 2020, 8861251.
  32. Liu, Y.; Weick, J.P.; Liu, H.; Krencik, R.; Zhang, X.; Ma, L.; Zhou, G.M.; Ayala, M.; Zhang, S.C. Medial ganglionic eminence-like cells derived from human embryonic stem cells correct learning and memory deficits. Nat. Biotechnol 2013, 31, 440–447.
  33. Fujikawa, T.; Oh, S.H.; Pi, L.; Hatch, H.M.; Shupe, T.; Petersen, B.E. Teratoma formation leads to failure of treatment for type I diabetes using embryonic stem cell-derived insulin-producing cells. Am. J. Pathol. 2005, 166, 1781–1791.
  34. Jin, X.; Lin, T.; Xu, Y. Stem Cell Therapy and Immunological Rejection in Animal Models. Curr. Mol. Pharmacol. 2016, 9, 284–288.
  35. Tolosa, L.; Pareja, E.; Gómez-Lechón, M.J. Clinical Application of Pluripotent Stem Cells: An Alternative Cell-Based Therapy for Treating Liver Diseases? Transplantation 2016, 100, 2548–2557.
  36. Israel, M.A.; Yuan, S.H.; Bardy, C.; Reyna, S.M.; Mu, Y.; Herrera, C.; Hefferan, M.P.; Van Gorp, S.; Nazor, K.L.; Boscolo, F.S. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 2012, 482, 216–220.
  37. Katsuda, T.; Tsuchiya, R.; Kosaka, N.; Yoshioka, Y.; Takagaki, K.; Oki, K.; Takeshita, F.; Sakai, Y.; Kuroda, M.; Ochiya, T. Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Sci. Rep. 2013, 3, 1–11.
  38. Hass, R.; Kasper, C.; Böhm, S.; Jacobs, R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun. Signal. 2011, 9, 1–14.
  39. Boutajangout, A.; Noorwali, A.; Atta, H.; Wisniewski, T. Human Umbilical Cord Stem Cell Xenografts Improve Cognitive Decline and Reduce the Amyloid Burden in a Mouse Model of Alzheimer’s Disease. Curr. Alzheimer Res. 2017, 14, 104–111.
  40. Lee, H.J.; Lee, J.K.; Lee, H.; Carter, J.E.; Chang, J.W.; Oh, W.; Yang, Y.S.; Suh, J.G.; Lee, B.H.; Jin, H.K.; et al. Human umbilical cord blood-derived mesenchymal stem cells improve neuropathology and cognitive impairment in an Alzheimer’s disease mouse model through modulation of neuroinflammation. Neurobiol. Aging 2012, 33, 588–602.
  41. Liu, X.Y.; Yang, L.P.; Zhao, L. Stem cell therapy for Alzheimer’s disease. World J. Stem Cells 2020, 12, 787–802.
  42. Yang, H.; Yang, H.; Xie, Z.; Wei, L.; Bi, J. Systemic transplantation of human umbilical cord derived mesenchymal stem cells-educated T regulatory cells improved the impaired cognition in AbetaPPswe/PS1dE9 transgenic mice. PLoS ONE 2013, 8, e69129.
  43. Ager, R.R.; Davis, J.L.; Agazaryan, A.; Benavente, F.; Poon, W.W.; LaFerla, F.M.; Blurton-Jones, M. Human neural stem cells improve cognition and promote synaptic growth in two complementary transgenic models of Alzheimer’s disease and neuronal loss. Hippocampus 2015, 25, 813–826.
  44. Lee, I.-S.; Jung, K.; Kim, I.-S.; Lee, H.; Kim, M.; Yun, S.; Hwang, K.; Shin, J.E.; Park, K.I. Human neural stem cells alleviate Alzheimer-like pathology in a mouse model. Mol. Neurodegener. 2015, 10, 1–16.
  45. Yang, H.; Xie, Z.H.; Wei, L.F.; Yang, H.N.; Yang, S.N.; Zhu, Z.Y.; Wang, P.; Zhao, C.P.; Bi, J.Z. Human umbilical cord mesenchymal stem cell-derived neuron-like cells rescue memory deficits and reduce amyloid-beta deposition in an AβPP/PS1 transgenic mouse model. Stem Cell Res. Ther. 2013, 4, 1–14.
  46. Kim, H.J.; Seo, S.W.; Chang, J.W.; Lee, J.I.; Kim, C.H.; Chin, J.; Choi, S.J.; Kwon, H.; Yun, H.J.; Lee, J.M. Stereotactic brain injection of human umbilical cord blood mesenchymal stem cells in patients with Alzheimer’s disease dementia: A phase 1 clinical trial. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2015, 1, 95–102.
More
Information
Subjects: Cell Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , , , , ,
View Times: 420
Entry Collection: Neurodegeneration
Revisions: 2 times (View History)
Update Date: 15 Nov 2022
1000/1000
Video Production Service