Document Type : Original Article

Authors

1 Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran

2 Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran

3 Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran

4 Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR), Mashhad, Iran

5 Private Veterinary Practitioner, Mashhad, Iran

Abstract

Spinal cord injury (SCI) presents challenging and unpredictable neurological recovery. During inflammatory conditions, the amount of serum albumin and nutrition consumption decreases. Currently, it is proposed to measure serum albumin and glucose content in human or animal subjects to predict the recovery rate and the efficiency of treatments following SCI. In this study, the effect of extra-cellular vesicles (EVs) from immortalized human adipose tissue-derived mesenchymal stem cells (hTERT-MSCs) equipped with the ectopic expression of the human indoleamine 2,3-dioxygenase-1 (IDO1) gene on serum albumin and glucose levels was investigated. After pre-clearing steps of 72-hr conditioned media, small EVs (sEVs) were isolated based on the ultra-filtration method. They were encapsulated with a chitosan-based hydrogel. Five experimental groups (female rats, N = 30, ~ 230 g) were considered, including SCI, sham, hydrogel, control green fluorescent protein (GFP)-EVs and IDO1-EVs. The 60.00 µL of hydrogel or hydrogels containing 100 µg sEVs from GFP or IDO1-EVs were locally injected immediately after SCI (laminectomy of the T10 vertebra and clip compression). After 8 weeks, non-fasting serum glucose and albumin levels were measured. The results indicated that the level of serum albumin in the animals received IDO1-EVs (3.52 ± 0.04) was increased in comparison with the SCI group (3.00 ± 0.94). Also, these animals indicated higher glucose levels in their serum (250.17 ± 69.61) in comparison with SCI ones (214 ± 45.34). Although these changes were not statistically significant, they could be considered as evidence for the beneficial effects of IDO1-EVs administration in the context of SCI to reduce hypoalbuminemia and improve energy consumption. More detailed experiments are required to confirm these results.

Keywords

Main Subjects

  1. Wertheim L, Edri R, Goldshmit Y, et al. Regenerating the injured spinal cord at the chronic phase by engineered iPSCs-derived 3D neuronal networks. Adv Sci (Weinh) 2022; 9(11): e2105694. doi: 10.1002/advs.202105694.
  2. Craig A, Perry KN, Guest R, et al. Prospective study of the occurrence of psychological disorders and comorbidities after spinal cord injury. Arch Phys Med Rehabil 2015; 96(8): 1426-1434.
  3. Zeng X, Xiang J, Dong L, et al. Advance in nutritional status and intervention after spinal cord injury. J Clin Nurs Res 2021; 5(6): 7-14.
  4. Bourguignon L, Vo AK, Tong B, et al. Natural progression of routine laboratory markers following spinal trauma: a longitudinal, multi-Cohort study. J Neurotrauma 2021; 38(15): 2151-2161.
  5. Tong B, Jutzeler CR, Cragg JJ, et al. Serum albumin predicts long-term neurological outcomes after acute spinal cord injury. Neurorehabil Neural Repair 2018; 32(1): 7-17.
  6. Brown SJ, Harrington GMB, Hulme CH, et al. A preliminary Cohort study assessing routine blood analyte levels and neurological outcome after spinal cord injury. J Neurotrauma 2020; 37(3): 466-480.
  7. Vo AK, Geisler F, Grassner L, et al. Serum albumin as a predictor of neurological recovery after spinal cord injury: a replication study. Spinal Cord 2021; 59(3): 282-290.
  8. Fanali G, di Masi A, Trezza V, et al. Human serum albumin: from bench to bedside. Mol Aspects Med 2012; 33(3): 209-290.
  9. Marcason W. Should albumin and prealbumin be used as indicators for malnutrition? J Acad Nutr Diet 2017; 117(7): 1144. doi: 10.1016/j.jand.2017.04.018.
  10. Gatta A, Verardo A, Bolognesi M. Hypoalbuminemia. Intern Emerg Med 2012; 7(Suppl 3): S193-S199.
  11. Kim SD, Melikian R, Ju KL, et al. Independent predictors of failure of nonoperative management of spinal epidural abscesses. Spine J 2014; 14(8): 1673-1679.
  12. Frisbie JH. Anemia and hypoalbuminemia of chronic spinal cord injury: prevalence and prognostic significance. Spinal Cord 2010; 48(7): 566-569.
  13. Khodabakhshi Rad A, Kazemi Mehrjerdi H, Pedram MS, et al. Clinical evaluation of the effect of methyl-prednisolone sodium succinate and meloxicam in experimental acute spinal cord injury. Iran J Vet Med 2023; 17(2):129-138. doi: 10.32598/IJVM. 17.2.1005246.
  14. Feng J, Zhang Y, Zhu Z, et al. Emerging exosomes and exosomal MiRNAs in spinal cord injury. Front Cell Dev Biol 2021; 9: 703989. doi: 10.3389/fcell.2021.703989.
  15. Sykova E, Cizkova D, Kubinova S. Mesenchymal stem cells in treatment of spinal cord injury and amyotrophic lateral sclerosis. Front Cell Dev Biol 2021; 9: 695900. doi: 10.3389/fcell.2021.695900.
  16. Navajas R, Corrales FJ, Paradela A. Serum exosome isolation by size-exclusion chromatography for the discovery and validation of preeclampsia-associated biomarkers. Methods Mol Biol 2019; 1959: 39-50.
  17. Ge W, Jiang J, Arp J, et al. Regulatory T-cell generation and kidney allograft tolerance induced by mesenchymal stem cells associated with indoleamine 2, 3-dioxygenase expression. Transplantation 2010; 90(12):1312-1320.
  18. Meireson A, Devos M, Brochez L. IDO expression in cancer: different compartment, different functionality? Front Immunol 2020; 11: 531491. doi: 10.3389/ fimmu.2020.531491.
  19. Proietti E, Pauwels RWM, de Vries AC, et al. Modulation of indoleamine 2, 3-dioxygenase 1 during inflammatory bowel disease activity in humans and mice. Int JTryptophan Res 2023; 16: 11786469231153109. doi: 10.1177/11786469231153109.
  20. Lukomska B, Stanaszek L, Zuba-Surma E, et al. Challenges and controversies in human mesenchymal stem cell therapy. Stem Cells Int 2019; 2019: 9628536. doi: 10.1155/2019/9628536.
  21. Zhang X, Jiang W, Lu Y, et al. Exosomes combined with biomaterials in the treatment of spinal cord injury. Front Bioeng Biotechnol 2023; 11: 1077825. doi: 10.3389/fbioe.2023.1077825.
  22. Hartjes TA, Mytnyk S, Jenster GW, et al. Extracellular vesicle quantification and characterization: common methods and emerging approaches. Bioengineering (Basel) 2019; 6(1): 7. doi: 10.3390/bioengineering 6010007.
  23. Mathieu M, Martin-Jaular L, Lavieu G, et al. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol 2019; 21(1): 9-17.
  24. Pluchino S, Smith JA. Explicating exosomes: reclassifying the rising stars of intercellular communication. Cell 2019; 177(2): 225-227.
  25. Zhang C, Deng R, Zhang G, et al. Therapeutic effect of exosomes derived from stem cells in spinal cord injury: a systematic review based on animal studies. Front Neurol 2022; 13: 847444. doi: 10.3389/fneur. 2022.847444.
  26. Théry C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the international society for extracellular vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018; 7(1): 1535750. doi: 10.1080/20013078.2018.1535750.
  27. Khalatbary AR. Stem cell-derived exosomes as a cell free therapy against spinal cord injury. Tissue Cell
    2021; 71: 101559. doi: 10.1016/j.tice.2021.101559.
  28. Wang K, Dong R, Tang J, et al. Exosomes laden self-healing injectable hydrogel enhances diabetic wound healing via regulating macrophage polarization to accelerate angiogenesis. Chem Eng J 2022; 430: 132664. doi: 10.1016/j.cej.2021.132664.
  29. Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy 2006; 8(4): 315-317.
  30. Börger V, Staubach S, Dittrich R, et al. Scaled isolation of mesenchymal stem/stromal cell-derived extra-cellular vesicles. Curr Protoc Cell Biol 2020; 55(1): e128. doi: 10.1002/cpsc.128.
  31. Leister I, Mittermayr R, Mattiassich G, et al. The effect of extracorporeal shock wave therapy in acute traumatic spinal cord injury on motor and sensory function within 6 months post-injury: a study protocol for a two-arm three-stage adaptive, prospective, multi-center, randomized, blinded, placebo-controlled clinical trial. Trials 2022; 23(1): 245. doi: 10.1186/ s13063-022-06161-8.
  32. Tátrai P, Szepesi Á, Matula Z, et al. Combined introduction of Bmi-1 and hTERT immortalizes human adipose tissue-derived stromal cells with low risk of transformation. Biochem Biophys Res Commun 2012; 422(1): 28-35.
  33. Haghighitalab A, Matin MM, Amin A, et al. Investigating the effects of IDO1, PTGS2, and TGF-β1 overexpression on immunomodulatory properties of hTERT-MSCs and their extracellular vesicles. Sci Rep 2021; 11: 7825. doi: 10.1038/s41598-021-87153-7.
  34. Haghighitalab A, Matin MM, Khakrah F, et al. Cost-effective strategies for depletion of endogenous extracellular vesicles from fetal bovine serum. J Cell Mol Res 2020; 11(2): 42-54.
  35. Naderi-Meshkin H, Andreas K, Matin MM, et al. Chitosan-based injectable hydrogel as a promising in situ forming scaffold for cartilage tissue engineering. Cell Biol Int 2014; 38(1): 72-84.
  36. Javdani M, Ghorbani R, Hashemnia M. Histo-pathological evaluation of spinal cord with experimental traumatic injury following implantation of a controlled released drug delivery system of chitosan hydrogel loaded with selenium nanoparticle. Biol Trace Elem Res 2021; 199(7): 2677-2686.
  37. Leister I, Linde LD, Vo AK, et al. Routine blood chemistry predicts functional recovery after traumatic spinal cord injury: a post hoc analysis. Neurorehabil Neural Repair 2021; 35(4): 321-333.
  38. Bernard F, Al-Tamimi YZ, Chatfield D, et al. Serum albumin level as a predictor of outcome in traumatic brain injury: potential for treatment. J Trauma 2008; 64(4): 872-875.
  39. Jin GX, Li L, Cui SQ, et al. Persistent hypoalbuminemia is a predictor of outcome in cervical spinal cord injury. Spine J 2014; 14(9): 1902-8.
  40. Yildirim T, Okutan O, Akpinar E, et al. Neuroprotective effects of high-dose human albumin against traumatic spinal cord injury in rats. Bratisl Lek Listy 2018; 119(2): 86-91.
  41. Wang ZG, He ZY, Liang S, et al. Comprehensive proteomic analysis of exosomes derived from human bone marrow, adipose tissue, and umbilical cord mesenchymal stem cells. Stem Cell Res Ther 2020; 11(1): 511. doi: 10.1186/s13287-020-02032-8.
  42. Rohde E, Pachler K, Gimona M. Manufacturing andcharacterization of extracellular vesicles from umbilical cord–derived mesenchymal stromal cells for clinical testing. Cytotherapy 2019; 21(6):
    581-592.
  43. Zeitler L, Murray PJ. IL4i1 and IDO1: oxidases that control a tryptophan metabolic nexus in cancer. J Biol Chem 2023; 299(6): 1 doi: 10.1016/ j.jbc.2023.104827.
  44. Murakami Y, Hoshi M, Imamura Y, et al. Remarkable role of indoleamine 2, 3-dioxygenase and tryptophan metabolites in infectious diseases: potential role in macrophage-mediated inflammatory diseases. Mediators Inflamm 2013; 2013: 391984. doi: 10.1155/ 2013/391984.
  45. Mándi Y, Vécsei L. The kynurenine system and immunoregulation. J Neural Transm (Vienna) 2012; 119(2): 197-209.
  46. Günther J, Fallarino F, Fuchs D, et al. Editorial: immunomodulatory roles of tryptophan metabolites in inflammation and cancer. Front Immunol 2020; 11: 1497. doi: 10.3389/fimmu.2020.01497.
  47. Liu JM, Deng HL, Chen XY, et al. Risk factors for surgical site infection after posterior lumbar spinal surgery. Spine (Phila Pa 1976) 2018; 43(10): 732-737.
  48. Scholpa NE, Schnellmann RG. Mitochondrial-based therapeutics for the treatment of spinal cord injury: mitochondrial biogenesis as a potential pharmacological target. J Pharmacol Exp Ther 2017; 363(3): 303-313.
  49. Cisternas P, Salazar P, Silva-Álvarez C, et al. Activation of Wnt signaling in cortical neurons enhances glucose utilization through glycolysis. J Biol Chem 2016; 291(50): 25950-25964.
  50. Giuliani G, Rosina M, Reggio A. Signaling pathways regulating the fate of fibro/adipogenic progenitors (FAPs) in skeletal muscle regeneration and disease. FEBS J 2022; 289(21): 6484-517.