Cell Therapy of Parkinson’s disease with Modified Neural Stem Cells: Explore the Possibilities

Chakraborty A and Diwan A

Published on: 1970-01-01

Abstract

Parkinson’s disease (PD) is characterized by movement disorders, slow speech, and many 21other additional non-motor function disabilities. PD results when a neurotransmitter in the substantia nigra (SN). There is currently no cure for PD available, only management options with DOPA supplementation. However, long-term use of DOPA therapies may result in significant adverse effects, like dyskinesia or motor neuron defects. A number of exciting treatment possibilities may result in near future. One of them is the regenerative cell therapy of PD. However, the main limitation is the availability of sufficient amounts of dopaminergic neuronal cells. Here, we discuss the possible modification method to generate sufficient, functional DOPA-producing cells for regenerative cell therapy of PD.

Keywords

Human neural stem cells; Melanocytes; Cell-Cell interaction; Dopamine; Parkinson’s disease

Introduction

Dopaminergic neurons of the midbrain are the main source of dopamine (DA) in the mammalian central nervous system. Their loss is associated with one of the most prominent human neurological disorders, Parkinson's disease (PD) [1,2]. The disease generally appears at middle age e.g. 50-60, but early onset has also been noticed; it is recognized by slow movement, tremor and with difficulty in normal postures [3,4]. Additionally, depression, anxiety, cognitive decline with dementia, sleep abnormalities are all evident with PD [5]. The real cause of PD is still unknown, although gene abnormalities have been found in 10-15% of PD cases. In other cases toxin exposure an head injuries are found to be involved. In brief, most of the PD cases have no clear cause, hence described still today as an idiopathic disease [6-9].

The number of PD cases in the world is growing noticeably [10]. Furthermore, no such curative treatments are available yet for PD, except palliative treatments like DOPA supplementation as a precursor for DA synthesis [11]. However, longtime use of DOPA as a supplement may cause dyskinesia, motor neuron defects, etc. [12-15].

Therefore, in recent years, instead of DOPA supplementation cell therapy with DOPA-producing cells is being in consideration. Neural stem cells (NSCs), induced pluripotent stem cells (iPSCs), and also other DOPA-producing cells like melanocytes have been studied for using as a cell therapeutic regimen for PD treatment [16-19].

In the recent past there are a number of publications showing the logistics and benefits of doing cell therapy of PD [20-28]. Embryonic stem cells (ESCs) are reported to differentiate into dopaminergic (DA-ergic) neurons, in vitro, using various protocols [29-31]. However the propensity to form teratomas along with ethical issues, limit their clinical uses [32,33]. Midbrain-derived hNSCs may be induced to differentiate into DA neurons, however, the lack of sufficient midbrain tissues is an issue. In addition, midbrain-derived hNSCs lose their proliferative and differentiation capacity in long-term cultures [32].

Human forebrain-derived hNSCs can be expanded in cultures for more than a year without losing their multipotency to differentiate into neurons and glial cells [32]. Thus, those cells can serve as suitable cell sources to provide sufficient number of candidate cells and to differentiate into DA neurons for transplantation uses in PD. More importantly, forebrain-derived hNSCs do not tend to form tumors when used for transplantation [34]. However, forebrain-derived hNSCs appear to have less potential to differentiate into functional DA neurons, which limits their therapeutic application in PD. Gene transfection technology has been reported to produce DA neurons in vitro [35], however there is a safety concern for clinical use of this genetically modified cell type. The therapeutic potential of NSC is broadly accepted by the scientific community but results from the clinical studies performed so far indicate that further studies are needed in order to improve their therapeutic efficacy. However, the in vitro generated iPSC-NSC, although similar to primary neuronal cell lines do not truly recapitulate the phenotype and function of endogenous NSC and retain the risk of tumor formation [36]. In addition, the reprogramming and differentiation processes are lengthy and costly under GMP conditions [36] NewNSC sources and a better understanding of the neurobiology of the cells will surely help in the ambitious quest to 75 repair the brain using cell-based therapies [36].

We have tried previously with hNSCs, primary human neural stem cells derived from the NIH approved H9 (WA09) human (female origin) embryonic stem cells; purchased from ThermoFisher (Rockford, IL, USA) to examine their potential to produce neurons, growth potential and functional differentiation capacity.

However, the problem still remains with their availability in sufficient amount needed for PD cell therapy. This article emphasizes the improvement of the cell quality and quantity, herein hNSCs, by a non-genetic modification method, i.e. cell-cell communication, before transplantation in the PD brain. By cell-cell communication, we mean, either using a cell-cell-co-culturing system and/or cell-cell fusion. However, at present we are focusing mainly on cell-cell co-culture system, not cell fusion. However, we planned to use a 2D cell culture system as our goal to monitor the growth and dopamine secretion by the hNSCs in presence of partner cells hMCs. Similar methods and results have been reported previously using Fibroblasts and HeLa cells [37].

Cell Therapy

Transplantation of a new NSC cell population can restore the lost nigrostriatal DA transmission in PD patients [38]. Various cell lines including genetically modified cells, iPSCs (induced-pluripotent-stem-cells), have been choices for transplantation in PD patients [39,40]. However, there are many issues like possible development of teratomas, ethical issues, cost and time effectiveness of the process makes the choice limited [41-44].

Embryonic stem cells (ESCs) though having the potential to be differentiated into dopamine producing cells in vitro [39,40,44], possess the issues as mentioned above to limit their use in clinics [41,42]. Although midbrain-derived neural stem cells (mNSCs) exhibit the potential to differentiate into Dopaminergic neurons (DA-ergic), their clinical application is restricted due to their low growth potential and multi-potency for differentiation in culture [41]. On the other hand, fetal forebrain neural stem cells (NSCs) can be cultured for a long time without losing their ability to be differentiated into neurons and glial cells [41].

Another neural origin cell-type, melanocytes that are mainly responsible for skin melanin formation from tyrosine, using tyrosinase via DOPA. Therefore, these cells were also proposed for transplantation in PD patients for supplying dopamine in their brain [17-19]. In fact, tyrosinase was found to reverse the PD symptoms in the brain of tyrosine hydroxylase (TH)-null mice [18,45]. Therefore, a therapeutic potential of melanocyte transplantation in the PD brain can be understood. However, melanocytes do not have other DOPA metabolizing enzymes, like Dopamine Transporter (DAT), Mono-amino-oxidase B (MAO-B), and Catecholamine Transferase (COMT), which are needed to catabolize the excess dopamine from the synaptic cleft. The excess dopamine can cause the toxic effects like dyskinesia and motor neuron defect, etc. [12-15,46,47]. Generation of Tyrosine Hydroxylase-positive neurons from human Embryonic Stem Cells were shown after co-culture with cellular substrates and exposure to Glial cell line-derived neurotrophic factor (GDNF) [48,49]. These modified cells also express DAT (Dopamine Transporter), COMT, and MAO-B that together can maintain the physiological level of dopamine in the synaptic cleft [50,51]. Therefore, hNSCs can be considered as a better choice over the other cell types for cell transplantation therapy of PD patients [52]. However, slow growth of hNSCs and senescence after a few passages hinders the ability to procure enough cells needed for transplantation. We are trying to test whether the hNSCs can be modified by co-culturing with melanocytes for their increased growth potential, survival length along and their DA-ergic ability. In brief, for cell replacement therapy of PD, our goal is to select or modify any existing potential cell type for their moderate growth potential, differentiation capacity, axon production and DA-ergic ability. Lastly, the cell should metabolize any excess dopamine from the synaptic cleft to prevent the possibilities of hallucinations, motor neuron defects and other side effects.

Cell-Cell Interaction, a Method of Cell Modification

Cell-cell interaction has been reported to modify each cell in the interaction [53]. This process of communications in between two cells is well evident in somatic cell biology in vivo and in vitro, and in cancer cell biology, as well [54,55]. Cancer cells can interact with their surrounding macrophages, and that communication results in either tumorsuppression or progression depending on the type Macrophages interacting with the cancer cells [56-61]. Cancer-associated fibroblasts can also promote growth and progression of the cancer cells [62]. Furthermore, cancer cells on interaction with endothelial cells can elongate blood vessels [63]. Overall, cell-cell communications mutually can alter the phenotypes and genotypes of each other [64].

Intercellular Communication as a Method for Neural Cell Modification

Like immune cell synapses, the classical neuronal synapses are involved in cell-cell adhesion and membrane association. Studies have shown that the release of serotonin (5-HT) in mice as well as in humans results from the interaction of dendritic cells (DC) and the naive T-cells [65,66] Together, many biological functions such as, embryonic development, neurotransmission, wound healing, inflammation, etc. are all shown to result from cell-cell interaction [60,67].

Cell–Cell Interactions for Neuron Formation

Neurons and axons interact with each other in normal neuro-circuitry pathways [68]. Under ischemic conditions, microglial phagocytosis of apoptotic neurons were induced by remodeling fractalkine receptor (CX3CL1/CX3CR1) mediated signaling [69]. NSPCs (neuronal stem progenitor cells) after activation migrate to the ischemic lesion and proliferate there [70]. Many other incidences of similar of cell–cell communications under stress were also reported. In glial cells, metabolic deficits in axonal energy are related to neurodegenerative diseases, like PD and AD [71]. Oligodendrocytes release lactate to neurons through MCT1 protein (monocarboxylate transporter1), and it’s dysfunction causes neuronal/axonal damage [72]. In fact, deficiency of MCT1 leads to a disruption of the trophic support to neurons (axons) found in an ALS (amyotrophic lateral sclerosis) mouse model as well as in human subjects [73].

Cell-Cell Interaction Can Effect Dopamine Production

Dopamine is the main neurotransmitter released by NSCs in brain, responsible for movement, endocrine regulation and cardiovascular function, as well as in maintaining the cognitive functions [74,75]. Further, dopamine is the main precursor of adrenaline and noradrenaline hormone releases in the nerve periphery [76]. Leukocytes, which are abundant in surrounding NSCs, release dopamine which may act as an autocrine and paracrine immune modulator [77]. The synthesis and storage of dopamine in monocyte-derived 168         dendritic cells (Mo-DCs) can be enhanced by cAMP producing cells [78]. Likewise, DA-ergic cells can increase cAMP levels in CD41+ T cells [CD4+T cells recognize peptides presented on MHC class II molecules, which are found on antigen presenting cells (APCs)], indicating a possible cross-talk are there for mutual benefit.

Partner Cell Selection for Interaction with hNSCs

The followings are the cells that can be considered for interaction with hNSCs, but the       question is which one should be the best choice for the purpose. Table 1 describes the different cells and their merits and demerits in using for modifying the NSCs.

Table 1: Cells with their Merits and Demerits in using for Modifying NSCs.

Human Embryonic Stem Cells (hESCs)

Neural Progenitors Cells (NPCs)

Bone Marrow Derived Adult Progenitor Cells

Melanocytes

 

ES can modify the growth potential of other cells [79]

 

NPCs are multipotent [84]

 

Bone marrow derived adult progenitor cells can trans-differentiate and generate neurons in vivo [87-90]

Melanocytes are originated from neural-crest, and committed to form skin melanin [90,91]

hES cells can modify the growth potential of hNSCs by cell-cell interaction [80]

 

NPCs can migrate, differentiate into neurons and can make synaptic contacts with the local neurons [84,85]

Stem cells may not have originally the neurons but can extends a new neuron on fusion with the host neurons [88,89]

These cells express melanocortin receptor (MC1) and releases the second messenger, cAMP [91]

The possibilities of forming teratomas and ethical issues limits the use of hES cells [81-83]

Use of this cell for clinical purpose, however, is still in dispute [85-87]

 

 

cAMP and/or its inducers can increase dopa and dopamine production, in vivo and in vitro [92,93] 

 

 

 

Dopamine or its precursor dopa can increase the cAMP level through D1-receptor-mediated signaling [92,93]

 

 

 

Therefore, a cross-talk between the neural cells and melanocytes can be expected.

Our Approach

From the above information, Melanocytes can be considered possibly as a better partner cell type over others to modify the growth potential of NSCs. We are interested to study, in particular, whether melanocyte can influence neural cells for their proliferation, increased survival length, along with other inherent properties like, axon production, dopamine and growth factors BDNF/GDNF secretion.

We have studied cell–cell interaction by co-culturing human normal melanocytes (hNMCs) with human Neural Stem Cells (hNSCs) in vitro [94,95]. hNMCs primary normal human adult melanocytes, lightly pigmented, from male origin were purchased from ATCC, Manassas, VA, Cat# PCS-200-012). These cells produce and secrete melanin. The source and type of human Neural Stem Cells (hNSCs) are already mentioned above in Section 1 [94,95].

This co-culture method is quite similar to co-culturing cancer cells andendothelial cells, fibroblasts and immune cells, reported previously [37]. In brief, we used the 2D culture system according to the reference citations, which differs from 3D culturing system in many ways but still possesses several advantages [For comparison, see Ref: 96]. Although the 3D cultring system mimics mostly the in vivo condition, it cannot exchange nutrients and waste freely like 2D culture. Furthermore, 2D cell cultures are easier for environmental control, observation, imaging, measurement and eventual manipulation, if needed.

Documentation of cell growth, cell morphology and axon production at different time points were done. Expression levels of NSCs-specific proteins like, Nestin, Tubulin III (Tuj1) were studied using immuno-fluorescent microscopy. ELISAs were done for measuring the secretion of Dopamine, BDNF and GDNF by NSCs. In brief, co-culturing of hNSCs with hNMCs in vitro showed an increased growth potential of hNSC’s, survival length, and increased capability in producing dopamine, BDNF/GDNF as reported earlier [94,95].

Discussion

The human body is composed of different types of cells with specialized functions, and they do communicate with other cells from different organs [97, 98]. This communication occurs either through direct physical contact, receptor-ligand interaction, cellular junction, or via secreted materials like proteins, cytokines, metabolites, growth factors, etc. from the neighboring cells [99,100]. Interactions via extracellular vesicles are also a possible way of communication [101]. Furthermore, physical properties of the surrounding extracellular matrix,     hypoxia and energy level modulate cell-cell communication [102]. In humans, LO–LO (lipoxygenases) interactions positively modulate the biosynthesis of Lipoxin (LX). LXs are a local mediators produced from arachidonic acid responsible for distinct and potent biological roles in inflammation, infection, or injury [69,103].

Together, it is conceivable that cell-cell interaction can contribute for biological functions of the organism, such as, embryonic development, wound healing, neurotransmission, inflammation, etc. In the scenario of Parkinson’s disease (PD) where the loss of DA-ergic neural cells in substantia nigra (SN) region of the brain was evident, the replenishment of cellular loss with a modified neural cells, is expected to be a valid curative approach. Furthermore, we can predict from our co-culture experiment that melanocyte and NSCs can lead to an autocrine-paracrine communication for better growth of NSCs and their secretion of dopamine. Dopamine, in turn, may induce cAMP formation from melanocytes and that 2nd messenger can further induce the neighboring       melanocytes to produce DOPA, a precursor of dopamine produced by neural cells.

Acknowledgment

We acknowledge all of our staff members, scientists from AllExcel, Inc. Especially Ms. Beth Pond to support during the writing of this review by providing materials, and editing.

Authors Contribution

All the authors contributed equally.

Conflict of Interest

This research is supported by internal grant from AllExcel, Inc. The authors report no conflicts of interest.

References

  1. Cheng HC, Ulane CM, Burke RE Clinical progression in Parkinson’s disease and the neurobiology of axons. Ann Neurol. 2010; 67: 715?725.
  2. Patrick PM, HirschEC, Hunot  Understanding dopaminergic cell death pathways in parkinson disease. Neuron 2016; 90: 675-691.
  3. Kalia LV, Lang AE. Parkinson’s Disease. The Lancet. 2015; 386: 896-912.
  4. Khoo TK, Yarnall AJ, Duncan GW, Coleman S, O'Brien JT, Brooks DJ, et al. The spectrum of nonmotor symptomps in early Parkinson disease. Neurology 2013; 80: 276-281.
  5. Kalia LV, Lang AE. Parkinson’s disease. Lancet [Internet]. 2015; 386: 896-912.
  6. APDA: POTENTIAL CAUSES OF PARKINSON’S DISEASE.
  7. com. What causes parkinson's disease? 2022
  8. org. Understanding of Parkinsons Disease/cause.
  9. NIH National Institute on Aging (NIA): Parkinson’s Disease: Causes, Symptoms, and Treatment.
  10. Dorsey ER, Sherer T, Okun MS, Bloem BR. The emerging evidence of the parkinson pandemic. J Parkinsons Dis. 2018; 8: S3-S8.
  11. Dorszewska J, Prendecki M, Lianeri M, Kozubski W. Molecular effects of L-dopa therapy in Parkinson’s disease. Curr Genomics. 2014; 15: 11-17.
  12. Thanvi B, Lo N, Robinson T. Levodopa-induced dyskinesia in Parkinson’s disease: clinical features, pathogenesis, prevention and treatment. Postgrad Med J. 2007; 83: 384-388.
  13. Dyskinesia is a complication of long-term use of levodopa. 2022. 
  14. Liang TW. Tarsy D. Medical management of motor fluctuations and dyskinesia in Parkinson disease. Literature review current through.
  15. What causes dyskinesia?
  16. Han F, BarembergD, Gao I, Duan J, Lu X, Zhang N, Chen Development of stem cell-based therapy for Parkinson's disease. Transl Neurodegener 2015; 4: 38-39.  
  17. Mull AN, Zolekar A, Wang YC. Understanding melanocyte stem cells for disease modeling and regenerative medicine applications. Int J Mol Sci 2015; 16: 30458?30469.
  18. Asanuma M, Miyazaki I, Diaz-Corrales F, Higashi Y, Namba M, Ogawa N. Transplantation of melanocytes obtained from the skin ameliorates apomorphine-induced abnormal behavior in rodent hemi-parkinsonian models. PLoS ONE 2013; 8: e65983.
  19. Costin GE, Hearing VJ. Human skin pigmentation: melanocytes modulate skin color in response to stress. FASEB J. 2007; 21: 976-994.
  20. Björklund LM. Stem cell therapy for Parkinson's disease. Dialogues Clin Neurosci. 2004; 6: 303-311.
  21. Ambasudhan R, Dolatabadi N, Nutter A, Masliah E, Mckercher SR, Lipton SA. Potential for cell therapy in Parkinson's disease using genetically programmed human embryonic stem cell-derived neural progenitor cells. J Comp Neurol. 2014; 522: 2845-2856.
  22. Yasuhara T, Kameda M, Sasaki T, Tajiri N, Date I. Cell therapy for parkinson’s disease. Cell Transplantation. 2017; 26: 1551-1559.
  23. Parkinson’s Disease: Pathogenesis and Clinical Aspects. Stoker TB, Greenland JC (Editors). Codon Publications, Brisbane, Australia. ISBN: 978-0-9944381-6-4.
  24. Chakraborty A, Brauer S, Diwan A. Possible therapies of Parkinson’s disease: A review. J Clin Neurosc. 2020; 75; 1-4.
  25. Jang SE, Qiu L, Chan LL, Tan EK, Zeng L. Current status of stem Cell-Derived therapies for parkinson’s disease: From cell assessment and imaging modalities to clinical trials. Front Neurosci. 2020; 14: 558532.
  26. Kim TW, Koo SY, Studer L. Pluripotent stem cell therapies for parkinson disease: Present challenges and future opportunities. Front Cell Dev Biol. 2020; 8: 729.
  27. Zhaohui Liu, Cheung HH. Stem Cell-Based therapies for parkinson disease. Int J Mol Sci. 2020; 21: 8060.
  28. Sancho-Bielsa FJ. Parkinson's disease: Present and future of cell therapy. Neurology Perspectives. 2022; 2: 558-S68.
  29. Kirkeby A, Grealish S, Wolf DA, Nelander J, Wood J, Lundblad M, et al. Generation of regionally specified neural progenitors and functional neurons from human embryonic stem cells under defined conditions. Cell Rep. 2012; 1:703-714.
  30. Yang F, Liu Y, Tu J, Wan J, Zhang J, Wu B, et al. Activated astrocytes enhance the dopaminergic differentiation of stem cells and promote brain repair through bFGF. Nat Commun. 2014; 5: 5627.
  31. Lim MS, Shin MS, Lee SY, Minn YK, Hoh JK, Cho YH, al. Noggin over-expressing mouse embryonic fibroblasts and ms5 stromal cells enhance directed differentiation of dopaminergic neurons from human embryonic stem cells. PLoS ONE. 2015; 10: e0138460.
  32. Christophersen NS, Meijer X, Jorgensen JR, Englund U, Gronborg M, Seiger A, et al. Induction of dopaminergic neurons from growth factor expanded neural stem/progenitor cell cultures derived from human first trimester forebrain. Brain Res Bull. 2006; 70: 457-466.
  33. Knoepfler PS. Deconstructing stem cell tumorigenicity: a roadmap to safe regenerative medicine. Stem Cells. 2009; 27: 1050-1056.
  34. Daadi MM, Grueter BA, Malenka RC, Redmond DE Jr, Steinberg GK. Dopaminergic neurons from midbrain-specified human embryonic stem cell-derived neural stem cells engrafted in a monkey model of parkinson’s disease. PLoS ONE. 2012; 7: e41120.
  35. Andersson EK, Irvin DK, Ahlsiö J, Parmar M. Ngn2 and nurr1 act in synergy to induce midbrain dopaminergic neurons from expanded neural stem and progenitor cells. Exp Cell Res.2007; 313: 1172-1180.
  36. Fernandez-Muñoz B, Garcia-Delgado AB, Arribas-Arribas B, Sanchez-Pernaute R. Human neural stem cells for Cell-Based medicinal products. Cells. 2021; 10: 2377-2403.
  37. Delinasios JG, Angeli F, Koumakis G, Kumar S, Kang W-H, Sica G, et. al. Proliferating Fibroblasts and HeLa Cells Co-cultured In Vitro Reciprocally Influence Growth Patterns, Protein Expression, Chromatin Features and Cell Survival. Anticancer Research. 2015; 35: 1881-1916.
  38. Gaillard A, Jaber J. Rewiring the brain with cell transplantation in parkinson’s disease. Trends in Neurosciences. 2011; 34: 124-133.
  39. Christophersen NS, Meijer X, Jorgensen JR, Englund U, Gronborg M, Seiger A, et. al. Induction of dopaminergic neurons from growth factor expanded neural stem/progenitor cell cultures derived from human first trimester forebrain. Brain Res Bull. 2006; 70: 457-466.
  40. Knoepfler PS. Deconstructing stem cell tumorigenicity: a roadmap to safe regenerative medicine. Stem Cells. 2009; 27: 1050-1056.
  41. Medvedev SP, Shevchenko AI, and Zakian SM. Induced Pluripotent Stem Cells: Problems and Advantages when Applying them in Regenerative Medicine. Acta Naturae. 2010; 2: 18-28.
  42. Lim MS, Shin MS, Lee SY, Minn YK, Hoh JK, Cho YH, et al. Noggin over-expressing mouse embryonic fibroblasts and ms5 stromal cells enhance directed differentiation of dopaminergic neurons from human embryonic stem cells. PLoS ONE. 2015; 10: e0138460.
  43. Rios M, Habecker B, Sasaoka T, Eisenhofer G, Tian H, Landis S, et al. Catecholamine synthesis is mediated by tyrosinase in the absence of tyrosine hydroxylase. J Neurosci. 1999; 19: 3519-3526.
  44. Zhang M. Two-step production of monoamines in monoenzymatic cells in the spinal cord: a different control strategy of neurotransmitter supply? Neural Regen Res. 2016; 11: 1904-1909.
  45. Keber U, Klietz M, Carlsson T, Oertel WH, Weihe E, Schäfer MK, et al. Striatal tyrosine hydroxylase-positive neurons are associated with L-DOPA-induced dyskinesia in hemiparkinsonian mice. Neuroscience. 2015; 298: 302-317. 
  46. Suzuki S, Kawamata J, Iwahara N, Matsumura A, Hisahara S, Matsushita T, et al. Intravenous mesenchymal stem cell administration exhibits therapeutic effects against 6-hydroxydopamine-induced dopaminergic neurodegeneration and glial activation in rats. Neurosci Lett.2015; 584: 276-281.
  47. Buytaert-Hoefen KA, Alvarez E, Freed CR. First published: 23 December 2008.
  48. Colette Daubner S, Le T, Wang Tyrosine Hydroxylase and Regulation of Dopamine Synthesis. Arch Biochem Biophys. 2011; 508: 1-12.
  49. German CL, Baladi MG, McFadden LM, Hanson GR, Fleckenstein AE. Regulation of the dopamine and vesicular monoamine transporters: pharmacological targets and implications for disease. Pharmacol Rev. 2015; 67: 1005-1024.
  50. Chakraborty A, Diwan A. Selection of Cells for Parkinson’s Disease Cell-Therapy. Int J Stem Cell Res Ther. 2019; 6: 063.
  51. Almet AA, Cang Z, Jin S, Nie Q, et al. The landscape of cell–cell communication through single-cell transcriptomics. Current Opinion in Systems Biology. 2021; 26: 12-23
  52. Gloushankova NA, Rubtsova SN, Zhitnyak IY. Cadherin-mediated cell-cell interactions in normal and cancer cells. Tissue Barriers. 2017; 5: e1356900.
  53. Kami?ska K, Szczylik C, Bielecka ZF, Bartnik E, Porta C, Lian F, Czarnecka AM. The role of the cell-cell interactions in cancer progression. J Cell Mol Med. 2015; 19: 283-296.
  54. Gajewski T, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013; 14: 1014-1022.
  55. Noy R and Pollard JW. Tumor-Associated Macrophages: From Mechanisms to Therapy. Immunity. 2014; 41: 49-62.
  56. MantovaniA, Sozzani S, Locati M, Allavena P, Sica A Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends in Immunology. 2002; 23: 549-555. 
  57. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018; 24: 541-550.
  58. Nishida-Aoki N, Gujral TS. Emerging approaches to study cell-cell interactions in tumor microenvironment. Oncotarget. 2019; 10: 785-797.
  59. Pawelek J, Chakraborty A. Fusion of tumor cells with bone marrow-derived cells: a unifying explanation for metastasis. Nat Rev Cancer. 2008; 8: 377-386.
  60. Xing F, Saidou J, Watabe K. Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front Biosci (Landmark Ed). 2010; 15: 166-179.
  61. RiveraLB, Bergers Intertwined regulation of angiogenesis and immunity by myeloid cells. Trends Immunol. 2015; 36: 240-249. 
  62. Semenza GL, Ruvolo Introduction to tumor microenvironment regulation of cancer cell survival, metastasis, inflammation, and immune surveillance. Biochim Biophys Acta. 2016; 1863: 379-381.
  63. O'Connell PJ, Wang X, Leon-Ponte M, Griffiths C, Pingle SC, Ahern GP. A novel form of immune signaling revealed by transmission of the inflammatory mediator serotonin between dendritic cells and T cells. Blood. 2006; 107: 1010-1017.
  64. Pacheco H, Martinez-Navío JM, Climent N, Ciruela F, Gatell JM, Gallart T, et al. Glutamate released by dendritic cells as a novel modulator of T cell activation. J Immunol. 2006; 177: 6695-704.
  65. Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med. 2014; 6: 265sr6.
  66. Zou Y. Does planar cell polarity signaling steer growth cones? Curr Top Dev Biol.2012; 101: 141-160.
  67. Cardona A, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra IM, et al. Control of microglial neurotoxicity by the fractalkine receptor. Nature Neuroscience. 2006; 9: 917-924. 
  68. Zhang RL, Zhang ZG, Chopp M. Ischemic stroke and neurogenesis in the subventricular zone. Neuropharmacology. 2008; 55: 345-52.
  69. Rõlova T, Lehtonen S, Goldsteins G, Kettunen P, Koistinaho J. Metabolic and immune dysfunction of glia in neurodegenerative disorders: Focus on iPSC models. Stem Cells. 2021; 39: 256-265.
  70. Morrison BM, Lee Y, Rothstein JD. Oligodendroglia: metabolic supporters of axons. Trends Cell Biol. 2013; 23: 644?651. 
  71. Kang SH, Fukaya M, Yang JK, Rothstein JD, Bergles DE. NG2+ CNS glial progenitors remain committed to the oligodendrocyte lineage in postnatal life and following neurodegeneration. 2010; 68: 668-681. 
  72. Juárez Olguín H, Calderón Guzmán D, Hernández García E, Barragán Mejía G. The Role of Dopamine and Its Dysfunction as a Consequence of Oxidative Stress. Oxid Med Cell Longev. 2016; 9730467.
  73. Ott T and Nieder A. Dopamine and Cognitive Control in Prefrontal Cortex. Trends in Cognitive Sciences. (Cells Press Reviews). 2019; 23: 213-234.
  74. Litwack G. Chapter 11 - Hormones of the adrenal medulla, Editor(s): Gerald Litwack, Hormones (Fourth Edition), Academic Press, 2022, Pages 269-285, ISBN 9780323902625.
  75. Nakano K, Higashi T, Takagi R, Hashimoto K, Tanaka Y, Matsushita S. Dopamine released by dendritic cells polarizes Th2 differentiation. International Immunology. 2009; 21: 645-654.
  76. Nakano K, Takehiro Higashi T, Rie Takagi, Kumiko Hashimoto, Yoshiya Tanaka, Sho Matsushita. Dopamine released by dendritic cells polarizes Th2 differentiation. International Immunology. 21, issue 6, Pages 645-654.
  77. Vazin T, Freed WJ. Human embryonic stem cells: derivation, culture, and differentiation: a review. Restor Neurol Neurosci. 2010; 28: 589-603.
  78. Daadi MM, Steinberg GK. Manufacturing neurons from human embryonic stem cells: biological and regulatory aspects to develop a safe cellular product for stroke cell therapy. Regen Med. 2009; 4: 251-263.
  79. Ben-Hur T, Idelson M, Khaner H, Pera M, Reinhartz E, Itzik A, et al. Transplantation of human embryonic stem cell-derived neural progenitors improves behavioral deficit in Parkinsonian rats. Stem Cells. 2004; 22: 1246-1255.
  80. SchulzTC, Noggle SA, Palmarini GM, Weiler DA, Lyons IG, Pensa KA,  al. Differentiation of human embryonic stem cells to dopaminergic neurons in serum-free suspension culture. Stem Cells. 2004; 22: 1218-1238.
  81. Ramos-Zúñiga R, González-Pérez O, Macías-Ornelas A, Capilla-González V, Quiñones-Hinojosa A, "Ethical Implications in the Use of Embryonic and Adult Neural Stem Cells". Stem Cells Int. 2012; 1-7. 
  82. Emsley JG, Mitchell BD, Kempermann G, and Macklis JD. Adult neurogenesis and repair of the adult CNS with neural progenitors, precursors, and stem cells. Prog Neurobiol.2005; 75: 321-341.
  83. Lie DC, Song H, Colamarino SA, Ming GL, and Gage FH. Neurogenesis in the adult brain: new strategies for central nervous system diseases. Annu Rev Pharmacol Toxicol. 2004; 44: 399-421.
  84. Jain K. Cell therapy for CNS trauma. Molecular biotechnology. 2009; 42: 367-376.
  85. Sayles M, Jain M, Barker RA. The cellular repair of the brain in Parkinson’s disease--past, present and future.  Immunol.2004; 12: 321-342.
  86. Brazelton TR, Rossi FM, Keshet GI, Blau HM. From marrow to brain: expression of neuronal phenotypes in adult mice. Science. 2000; 290: 1775-1779.
  87. Cogle CR, Yachnis AT, Laywell ED, Zander DS, Wingard JR, Steindler DA, et al. Bone marrow transdifferentiation in brain after transplantation: a retrospective study.  2004; 363: 1432-1437.
  88. Terada N, Hamazaki T, Oka M, Hoki M, Mastalerz DM, Nakano Y, et al. Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion. Nature. 2002; 416: 542-545.
  89. Ying QL, Nichols J, Evans EP, and Smith AG. Changing potency by spontaneous fusion. Nature. 2002; 416: 545-548.
  90. Cichorek M, Wachulska M, Stasiewicz A, & Tymi?ska A. Skin melanocytes: Biology and development. Advances in Dermatology and Allergology. 2013; 30: 30-41. 
  91. Carlson JA, Linette GP, Aplin A, Ng B, Slominski A. Melanocyte receptors: clinical implications and therapeutic relevance. Dermatol Clin. 2007; 25: 541-57.
  92. Vaquero CF, Pignatelli A, Partida GJ, Ishida AT. A dopamine- and protein kinase A-dependent mechanism for network adaptation in retinal ganglion cells. J Neurosci. 2001; 21: 8624-8635.
  93. Rangel-Barajas C, Coronel I, Floran B. Dopamine receptors and neurodegeneration. Aging Dis. 2015; 6: 349-368.
  94. Chakraborty A, Diwan A. Autocrine and paracrine stimulation of dopamine secretion by human neural stem cells: Role BDNF and GDNF. Neurol Neurosci Rep. 2020; 3: 1-6.
  95. Chakraborty A and Diwan A. Coculturing NSCs with melanocyte increased its dopamine and neural factor secretion. Acta Scientific Neurology. 2021; 4: 70-78.
  96. Kapa?czyn?ska M, Kolenda T, Przyby?a W, Zaja?czkowska M, Teresiak A, Filas V, et al. 2D and 3D cell cultures – a comparison of different types of cancer cell cultures Arch Med Sci. 2018; 14, 4: 910-919
  97. Alberts B, Johnson A, Lewis J, et al. Molecular Biology of the Cell. 4th edition. New York: Garland Science; 2002. General Principles of Cell Communication.
  98. Bianconi E, Piovesan A, Facchin F, Beraudi A, Casadei R, Frabetti F. et al. An estimation of the number of cells in the human body. Annals of Human Biology. 2013; 40: 463-471.
  99. West AP, Khoury-Hanold W, Staron M, Tal MC, Pineda CM, Lang SM, et al. Mitochondrial DNA stress primes the antiviral innate immune response. 2015; 520: 553-557.
  100. Lyssiotis CA, Kimmelman AC. Metabolic Interactions in the Tumor Microenvironment. Trends Cell Biol. 2017; 27: 863-875.
  101. Becker A, Thakur BK, Weiss JM, Kim H, Peinado H, Lyden D. Extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell. 2016; 30: 836-848.
  102. Pickup MW, Mouw JK, Weaver VM. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 2014; 15: 1243-1253.
  103. Serhan CN, Levy BD. LIPID MEDIATORS, Lipoxins. Editor(s): Geoffrey J. Laurent, Steven D. Shapiro. Encyclopedia of Respiratory Medicine, Academic Press. 2006; 594-599.