We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Priming mesenchymal stromal cells with neurotrophic factors boosts the neuro-regenerative potential of their secretome

    Prajakta Teli

    Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India

    Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, 412115, India

    ,
    Aditya Nachanekar

    Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India

    Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, 412115, India

    ,
    Vaijayanti Kale

    Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India

    Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, 412115, India

    &
    Anuradha Vaidya

    *Author for correspondence:

    E-mail Address: anuradha.vaidya@ssbs.edu.in

    Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India

    Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, 412115, India

    Published Online:https://doi.org/10.2217/rme-2022-0201

    Aim: To explore the neuroprotective potential of the secretome (conditioned medium, CM) derived from neurotrophic factors-primed mesenchymal stromal cells (MSCs; primed CM) using an endoplasmic reticulum (ER) stress-induced in vitro model system. Methods: Establishment of ER-stressed in vitro model, immunofluorescence microscopy, real-time PCR, western blot. Results: Exposure of ER-stressed Neuro-2a cells to the primed-CM significantly restored the neurite outgrowth parameters and improved the expression of neuronal markers like Tubb3 and Map2a in them compared with the naive CM. Primed CM also suppressed the induction of apoptotic markers Bax and Sirt1, inflammatory markers Cox2 and NF-κB, and stress kinases such as p38 and SAPK/JNK in the stress-induced cells. Conclusion: The secretome from primed MSCs significantly restored ER stress-induced loss of neuro-regenesis.

    Graphical abstract

    Plain language summary

    Endoplasmic reticulum (ER) stress-mediated accumulation of misfolded protein is one of the causes involved in the onset of several neurodegenerative diseases (ND). Under physiological conditions, ER stress activates the unfolded protein response (UPR) that repairs the misfolded proteins. However, if the ER stress becomes chronic, the UPR fails to repair the misfolded proteins leading to disease conditions such as Parkinson's disease, Alzheimer's disease, multiple sclerosis, etc. Most in vitro systems are based on the infliction of acute ER stress on the target cells, which kills them, and thus, are not physiologically relevant models, as their neuro-regeneration is not possible. Here, we have developed a physiologically relevant in vitro model system, wherein we exposed Neuro-2a cells to an ER stress inducer which significantly affected their neuro-regenesis without killing them. These dysfunctional cells were then used to assess the effect of secretome (conditioned medium, CM) derived from mesenchymal stromal cells (MSCs) primed or not with neurotrophic factors. We found that priming of MSCs with neurotrophic factors enhances their neuroprotective potential. We demonstrate that when primed CM is given either as a single dose or in multiple doses, it restores neuro-regenesis and protects the stressed Neuro-2a cells from cell death. More importantly, the restoration of neuro-regenesis by primed CM is significantly higher as compared with the naive CM. These results clearly underscore the importance of priming the MSCs with neurotrophic factors for developing more effective therapeutic strategies to combat ND.

    Tweetable abstract

    Priming MSCs with neurotrophic factors boosts their neuroprotective potential. This strategy offers scope for developing competent cell-free therapies for the treatment of neurodegenerative diseases.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Esmaeili Y, Yarjanli Z, Pakniya F et al. Targeting autophagy, oxidative stress, and ER stress for neurodegenerative disease treatment. J. Control. Rel. 345, 147–175 (2022).
    • 2. Johri A, Beal MF. Mitochondrial dysfunction in neurodegenerative diseases. J. Pharmacol. Exp. Ther. 342(3), 619–630 (2012).
    • 3. Mattson MR. Calcium and neurodegeneration. Aging Cell 6(3), 337–350 (2007).
    • 4. Verkhratsky A, Toescu EC. Endoplasmic reticulum Ca(2+) homeostasis and neuronal death. J. Cell Mol. Med. 7(4), 351–361 (2003).
    • 5. Schrank S, Barrington N, Stutzmann GE. Calcium-Handling Defects and Neurodegenerative Disease. Cold Spring Harb. Perspect. Biol. 12(7), 1–25 (2020). • Discusses that calcium signaling is critical to neuronal function as it is the upstream underlying basis for the pathogenesis of neurodegenerative disease (ND).
    • 6. Sprenkle NT, Sims SG, Sánchez CL, Meares GP. Endoplasmic reticulum stress and inflammation in the central nervous system. Mol. Neurodegener. 12(1), 1–18 (2017). • Deliberated the mechanisms by which stress-induced inflammation contributes to the pathogenesis and progression of NDs.
    • 7. Walter P, Ron D. The unfolded protein response: from stress pathway to homeostatic regulation. Science 334(6059), 1081–1086 (2011).
    • 8. Lindholm D, Wootz H, Korhonen L. ER stress and neurodegenerative diseases. Cell Death Differ. 13(3), 385–392 (2006).
    • 9. Tanna T, Sachan V. Mesenchymal Stem Cells: Potential in Treatment of Neurodegenerative Diseases. Curr. Stem Cell Res. Ther. 9(6), 513–521 (2014).
    • 10. Sudjarwo S, Sudjarwo G, Koerniasari K. Protective effect of curcumin on lead acetate-induced testicular toxicity in Wistar rats. Res. Pharm. Sci. 12(5), 381 (2017).
    • 11. Yao P, Zhou L, Zhu L, Zhou B, Yu Q. Mesenchymal Stem Cells: A Potential Therapeutic Strategy for Neurodegenerative Diseases. Eur. Neuro. l83(3), 235–241 (2020).
    • 12. Wobst HJ, Mack KL, Brown DG, Brandon NJ, Shorter J. The clinical trial landscape in amyotrophic lateral sclerosis – past, present, and future. Med. Res. Rev. 40(4), 1352–1384 (2020).
    • 13. Zriek F, di Battista JA, Alaaeddine N. Mesenchymal Stromal Cell Secretome: Immunomodulation, Tissue Repair and Effects on Neurodegenerative Conditions. Curr. Stem Cell Res. Ther. 16(6), 656–669 (2021).
    • 14. Kean TJ, Lin P, Caplan AI, Dennis JE. MSCs: delivery routes and engraftment, cell-targeting strategies, and immune modulation. Stem Cells Int. 2013, 732–742 (2013).
    • 15. Lee S, Choi E, Cha MJ, Hwang KC. Cell adhesion and long-term survival of transplanted mesenchymal stem cells: a prerequisite for cell therapy. Oxid. Med. Cell Longev. 2015, 632902 (2015).
    • 16. Wu X, Jiang J, Gu Z, Zhang J, Chen Y, Liu X. Mesenchymal stromal cell therapies: immunomodulatory properties and clinical progress. Stem Cell Res. Ther. 11(1), 345 (2020).
    • 17. Teli P, Kale V, Vaidya A. Extracellular vesicles isolated from mesenchymal stromal cells primed with neurotrophic factors and signaling modifiers as potential therapeutics for neurodegenerative diseases. Curr. Res. Transl. Med. 69(2), 103286 (2021). •• Discusses priming as a strategy for harnessing the therapeutic potential of the mesenchymal stem cells (MSCs0 for promoting neuroregeneration.
    • 18. Pendse S, Kale V, Vaidya A. Extracellular Vesicles Isolated from Mesenchymal Stromal Cells Primed with Hypoxia: Novel Strategy in Regenerative Medicine. Curr. Stem Cell Res. Ther. 16(3), 243–261 (2021).
    • 19. Kale VP. Application of “primed” Mesenchymal Stromal Cells in Hematopoietic Stem Cell Transplantation: Current Status and Future Prospects. Stem Cells Dev. 28(22), 1473–1479 (2019).
    • 20. de Cássia Noronha N, Mizukami A, Caliári-Oliveira C et al. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res. Ther. 10(1), 131 (2019). • Reviews several strategies of priming MSCs to improve their therapeutic potential.
    • 21. Jalnapurkar S, Moirangthem RD, Singh S, Limaye L, Kale V. Microvesicles Secreted by Nitric Oxide-Primed Mesenchymal Stromal Cells Boost the Engraftment Potential of Hematopoietic Stem Cells. Stem Cells 37(1), 128–138 (2019).
    • 22. Budgude P, Kale V, Vaidya A. Pharmacological Inhibition of p38 MAPK Rejuvenates Bone Marrow Derived-Mesenchymal Stromal Cells and Boosts their Hematopoietic Stem Cell-Supportive Ability. Stem Cell Rev. Rep. 17(6), 2210–2222 (2021).
    • 23. Moirangthem RD, Singh S, Adsul A, Jalnapurkar S, Limaye L, Kale VP. Hypoxic niche-mediated regeneration of hematopoiesis in the engraftment window is dominantly affected by oxygen tension in the milieu. Stem Cells Dev. 24(20), 2423–2436 (2015).
    • 24. Pendse S, Kale V, Vaidya A. The Intercellular Communication Between Mesenchymal Stromal Cells and Hematopoietic Stem Cells Critically Depends on NF-κB Signalling in the Mesenchymal Stromal Cells. Stem Cell Rev. Rep. 18(7), 2458–2473 (2022).
    • 25. Teli P, Kale V, Vaidya A. Mesenchymal stromal cells-derived secretome protects Neuro-2a cells from oxidative stress-induced loss of neurogenesis. Exp. Neurol. 354, 114107 (2022). •• Demonstrates that the secretome derived from MSCs exerts a neuroprotective effect on oxidative stress-induced loss of neurogenesis.
    • 26. Lawson B, Brewer JW, Hendershot LM. Geldanamycin, an hsp90/GRP94-binding drug, induces increased transcription of endoplasmic reticulum (ER) chaperones via the ER stress pathway. J. Cell. Physiol. 174(2), 107–108 (1998). •• Shows that geldanamycin explicitly binds to Grp94, which modulates the expression Grp78 and Chop, thereby leading to the induction of endoplasmic reticulum (ER) stress.
    • 27. Talwadekar M, Fernandes S, Kale V, Limaye L. Valproic acid enhances the neural differentiation of human placenta derived-mesenchymal stem cells in vitro. J. Tissue Eng. Regen. Med. 11(11), 3111–3123 (2017). •• Shows that MSCs primed with neurotrophic factors differentiate into neuronal-like cells, thereby underscoring the importance of priming MSCs to enhance their propensity to differentiate into neuronal-like cells.
    • 28. Shihan MH, Novo SG, le Marchand SJ, Wang Y, Duncan MK. A simple method for quantitating confocal fluorescent images. Biochem. Biophys. Rep. 25, 100916 (2021).
    • 29. Duangjan C, Rangsinth P, Zhang S, Wink M, Tencomnao T. Anacardium Occidentale L. Leaf Extracts Protect Against Glutamate/H2O2-Induced Oxidative Toxicity and Induce Neurite Outgrowth: The Involvement of SIRT1/Nrf2 Signaling Pathway and Teneurin 4 Transmembrane Protein. Front. Pharmacol. 12, 627738 (2021).
    • 30. Kang S, Chen X, Gong S et al. Characteristic analyses of a neural differentiation model from iPSC-derived neuron according to morphology, physiology, and global gene expression pattern. Sci Rep. 7(1), 12233 (2017).
    • 31. Hosoi T, Hyoda K, Okuma Y, Nomura Y, Ozawa K. Geldanamycin induces CHOP expression through a 4-(2-aminoethyl)-benzenesulfonyl fluoride-responsive serine protease. Cell Res. 17(2), 184–186 (2007).
    • 32. Kawada K, Iekumo T, Saito R et al. Aberrant neuronal differentiation and inhibition of dendrite outgrowth resulting from endoplasmic reticulum stress. J. Neurosci. Res. 92(9), 1122–1133 (2014). •• Shows that ER stress induction in neuronal cells inhibits neuronal differentiation and neurite outgrowth formation, suggesting that ER stress induction leads to loss of neurogenesis.
    • 33. Hu H, Tian M, Ding C, Yu S. The C/EBP homologous protein (CHOP) transcription factor functions in endoplasmic reticulum stress-induced apoptosis and microbial infection. Front Immuno. l9, 3083 (2019).
    • 34. Chaudhari N, Talwar P, Parimisetty A, d'Hellencourt CL, Ravanan P. A molecular web: endoplasmic reticulum stress, inflammation, and oxidative stress. Front. Cell Neurosci. 8, 213 (2014).
    • 35. Kitamura M. Control of NF-κB and inflammation by the unfolded protein response. Int. Rev. Immunol. 30(1), 4–15 (2011).
    • 36. Fribley A, Zhang K, Kaufman RJ. Regulation of apoptosis by the unfolded protein response. Methods Mol. Biol. 559, 191–204 (2009).
    • 37. Liu L, Arun A, Ellis L, Peritore C, Donmez G. SIRT2 enhances 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced nigrostriatal damage via apoptotic pathway. Front. Aging Neurosci. 6, 184 (2014).
    • 38. Outeiro TF, Kontopoulos E, Altmann SM et al. Sirtuin 2 inhibitors rescue alpha-synuclein-mediated toxicity in models of Parkinson's disease. Science 317(5837), 516–519 (2007).
    • 39. Gao HM, Hong JS. Why neurodegenerative diseases are progressive: uncontrolled inflammation drives disease progression. Trends Immunol. 29(8), 357–365 (2008).
    • 40. Lin WC, Chuang YC, Chang YS et al. Endoplasmic reticulum stress stimulates p53 expression through NF-κB activation. PLOS ONE 7(7), e39120 (2012).
    • 41. Lee KM, Kang BS, Lee HL et al. Spinal NF-kB activation induces COX-2 upregulation and contributes to inflammatory pain hypersensitivity. Eur. J. Neurosci. 19(12), 3375–3381 (2004).
    • 42. Darling NJ, Cook SJ. The role of MAPK signalling pathways in the response to endoplasmic reticulum stress. Biochim. Biophys. Acta Mol. Cell Res. 1843(10), 2150–2163 (2014).
    • 43. Raza SS, Seth P, Khan MA. ‘Primed’ Mesenchymal Stem Cells: a Potential Novel Therapeutic for COVID19 Patients. Stem Cell Rev. Rep. 17(1), 153–162 (2021).
    • 44. Outeiro TF, Kontopoulos E, Altmann SM et al. Sirtuin 2 inhibitors rescue alpha-synuclein-mediated toxicity in models of Parkinson's disease. Science 317(5837), 516–519 (2007).
    • 45. Tabas I, Ron D. Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress. Nat. Cell Biol. 13(3), 184–190 (2011).
    • 46. Wright AL, Zinn R, Hohensinn B et al. Neuroinflammation and Neuronal Loss Precede Aβ Plaque Deposition in the hAPP-J20 Mouse Model of Alzheimer's Disease. PLOS ONE 8(4), e59586 (2013).
    • 47. Cordeiro MF, Guo L, Coxon KM et al. Imaging multiple phases of neurodegeneration: a novel approach to assessing cell death in vivo. Cell Death Dis. 1(1), e3 (2010).
    • 48. Chong WC, Shastri MD, Eri R. Molecular Sciences Endoplasmic Reticulum Stress and Oxidative Stress: A Vicious Nexus Implicated in Bowel Disease Pathophysiology. Int. J. Mol. Sci. 18(4), 771 (2017).
    • 49. Malhotra JD, Kaufman RJ. Endoplasmic reticulum stress and oxidative stress: a vicious cycle or a double-edged sword? Antioxid. Redox Signal. 9(12), 2277–2293 (2007).
    • 50. Lindholm D, Korhonen L, Eriksson O, Kõks S. Recent insights into the role of unfolded protein response in ER stress in health and disease. Front. Cell Dev. Biol. 5, 48 (2017).
    • 51. Matus S, Glimcher LH, Hetz C. Protein folding stress in neurodegenerative diseases: a glimpse into the ER. Curr. Opin. Cell Biol. l23(2), 239–252 (2011).
    • 52. Tam AB, Mercado EL, Hoffmann A, Niwa M. ER Stress Activates NF-κB by Integrating Functions of Basal IKK Activity, IRE1 and PERK. PLoS One. 7(10), e45078 (2012).
    • 53. Bartels A, Leenders K. Cyclooxygenase and neuroinflammation in Parkinson's disease neurodegeneration. Curr. Neuropharmacol. 8(1), 62–68 (2010).
    • 54. Kaltschmidt B, Linker RA, Deng J, Kaltschmidt C. Cyclooxygenase-2 is a neuronal target gene of NF-κb. BMC Mol. Biol. 3, 16 (2002).
    • 55. Vukicévić V, Qin N, Balyura M et al. Valproic acid enhances neuronal differentiation of sympathoadrenal progenitor cells. Mol. Psychiatry 20(8), 941–950 (2015).
    • 56. Kim H, Zahir T, Tator CH, Shoichet MS. Effects of dibutyryl cyclic-AMP on survival and neuronal differentiation of neural stem/progenitor cells transplanted into spinal cord injured rats. PLOS ONE 6(6), e21744 (2011).
    • 57. Delcroix GJR, Curtis KM, Schiller PC, Montero-Menei CN. EGF and bFGF pre-treatment enhances neural specification and the response to neuronal commitment of MIAMI cells. Differentiation 80(4–5), 213–227 (2010).
    • 58. Yu IT, Park JY, Kim SH, Lee JS, Kim YS, Son H. Valproic acid promotes neuronal differentiation by induction of proneural factors in association with H4 acetylation. Neuropharmacology 56(2), 473–480 (2009).
    • 59. Zahir T, Chen YF, MacDonald JF, Leipzig N, Tator CH, Shoichet MS. Neural stem/progenitor cells differentiate in vitro to neurons by the combined action of dibutyryl cAMP and interferon-γ. Stem Cells Dev. 18(10), 1423–1432 (2009).
    • 60. Munoz-Perez E, Gonzalez-Pujana A, Igartua M, Santos-Vizcaino E, Hernandez RM. Mesenchymal stromal cell secretome for the treatment of immune-mediated inflammatory diseases: latest trends in isolation, content optimization and delivery avenues. Pharmaceutics 13(11), 1802 (2021).
    • 61. Pinho AG, Cibrão JR, Silva NA, Monteiro S, Salgado AJ. Cell secretome: basic insights and therapeutic opportunities for CNS disorders. Pharmaceuticals 13(2), 31 (2020).
    • 62. Varderidou-Minasian S, Lorenowicz MJ. Mesenchymal stromal/stem cell-derived extracellular vesicles in tissue repair: challenges and opportunities. Theranostics 10(13), 5979–5997 (2020).
    • 63. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9(6), 654–659 (2007).
    • 64. Kulkarni R, Bajaj M, Ghode S, Jalnapurkar S, Limaye L, Kale VP. Intercellular Transfer of Microvesicles from Young Mesenchymal Stromal Cells Rejuvenates Aged Murine Hematopoietic Stem Cells. Stem Cells 36(3), 420–433 (2018).
    • 65. Doyle L, Wang M. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 8(7), 727 (2019).
    • 66. Saint-Pol J, Gosselet F, Duban-Deweer S, Pottiez G, Karamanos Y. Targeting and Crossing the Blood-Brain Barrier with Extracellular Vesicles. Cells 9(4), 851 (2020).
    • 67. Vaquero J, Zurita M, Rico MA et al. Repeated subarachnoid administrations of autologous mesenchymal stromal cells supported in autologous plasma improve quality of life in patients suffering incomplete spinal cord injury. Cytotherapy. 19(3), 349–359 (2017).
    • 68. Zhao Y, Tang F, Xiao Z et al. Clinical study of neuroregen scaffold combined with human mesenchymal stem cells for the repair of chronic complete spinal cord injury. Cell Transplant. 26(5), 891–900 (2017).
    • 69. Dahbour S, Jamali F, Alhattab D et al. Mesenchymal stem cells and conditioned media in the treatment of multiple sclerosis patients: clinical, ophthalmological and radiological assessments of safety and efficacy. CNS Neurosci. Ther. 23(11), 866–874 (2017).
    • 70. Gowen A, Shahjin F, Chand S, Odegaard KE, Yelamanchili SV. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Challenges in Clinical Applications. Front. Cell Dev. Biol. 8, 149 (2020).
    • 71. Linares GR, Chiu CT, Scheuing L et al. Preconditioning mesenchymal stem cells with the mood stabilizers lithium and valproic acid enhances therapeutic efficacy in a mouse model of Huntington's disease. Exp. Neurol. 281, 81–92 (2016).
    • 72. Kim HW, Lee HS, Kang JM et al. Dual Effects of Human Placenta-Derived Neural Cells on Neuroprotection and the Inhibition of Neuroinflammation in a Rodent Model of Parkinson's Disease. Cell Transplant. 27(5), 814 (2018).
    • 73. Somoza R, Juri C, Baes M, Wyneken U, Rubio FJ. Intranigral transplantation of epigenetically induced BDNF-secreting human mesenchymal stem cells: implications for cell-based therapies in Parkinson's disease. Biol. Blood Marrow Transplant. 16(11), 1530–1540 (2010).
    • 74. Nouri F, Salehinejad P, Nematollahi-mahani SN, Kamarul T, Zarrindast MR, Sharifi AM. Deferoxamine Preconditioning of Neural-Like Cells Derived from Human Wharton's Jelly Mesenchymal Stem Cells as a Strategy to Promote Their Tolerance and Therapeutic Potential: an In Vitro Study. Cell. Mol. Neurobiol. 36(5), 689–700 (2016).