| Peer-Reviewed

Exploring the Secretome’s Biomarker and Analgesic Potential

Received: 1 April 2022    Accepted: 20 April 2022    Published: 10 May 2022
Views:       Downloads:
Abstract

Diagnostic and prognostic biomarkers of nerve injury and/or pain as well as improved pain therapeutics are needed, both on the battlefield to treat injured Service Members and in the civilian sector. Our previous research indicates that there are several differentially expressed (DE) extracellular vesicle-derived microRNAs (EV-miRNAs) isolated from rat plasma following spinal nerve ligation (SNL). As such, EV-miRNAs hold promise as biomarkers and therapeutic targets. The secretome contains biological mediators, including EVs, which are released into the extracellular space. In this study we focus on evaluating EV-non-coding RNAs (ncRNAs), examine effects of SNL on key protein expression in the prefrontal cortex (PFC), and test the secretome’s analgesic properties. To accomplish these goals, anesthetized male Sprague Dawley rats underwent SNL and nociceptive behavior measurements, plasma collection followed by EV RNA isolation, small RNA sequencing, and analysis. Expression of several key proteins in the PFC was determined by Wes/Jess analysis. The secretome bath was applied directly to the ligated nerve and the paw withdrawal threshold (PWT) was measured. We identified differences in several classes of ncRNAs such as piRNAs, snoRNAs, and snRNAs post-SNL. Levels of phosphorylated forms of P70S6K and ERK1 were increased in the dorsal PFC at 15 days post-SNL. Bath application of the secretome directly to the ligated nerve resulted in recovery of the reduced PWT (increased mechanical sensitivity) that is induced by SNL. Here, we have identified specific EV-ncRNAs that could contribute to the formation of pain. Furthermore, we have evaluated a novel product for analgesic efficacy that could function to exploit the underlying mechanisms that contribute to pain development, thus reducing acute pain. This is key in treating Service Members on the battlefield in order to prevent pain chronification.

Published in American Journal of Psychiatry and Neuroscience (Volume 10, Issue 2)
DOI 10.11648/j.ajpn.20221002.12
Page(s) 63-76
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2022. Published by Science Publishing Group

Keywords

Secretome, Nociception, Extracellular Vesicles, Analgesia, Spinal Nerve Ligation, Non-coding RNA

References
[1] Sosanya, N. M., et al., Identifying Plasma Derived Extracellular Vesicle (EV) Contained Biomarkers in the Development of Chronic Neuropathic Pain. J Pain, 2019.
[2] van Niel, G., G. D'Angelo, and G. Raposo, Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol, 2018. 19 (4): p. 213-228.
[3] Keshtkar, S., N. Azarpira, and M. H. Ghahremani, Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther, 2018. 9 (1): p. 63.
[4] Urabe, F., et al., Extracellular vesicles as biomarkers and therapeutic targets for cancer. Am J Physiol Cell Physiol, 2020. 318 (1): p. C29-C39.
[5] Hill, A. F., Extracellular Vesicles and Neurodegenerative Diseases. J Neurosci, 2019. 39 (47): p. 9269-9273.
[6] Harrell, C. R., et al., Mesenchymal Stem Cell-Derived Exosomes and Other Extracellular Vesicles as New Remedies in the Therapy of Inflammatory Diseases. Cells, 2019. 8 (12).
[7] Kharaziha, P., et al., Tumor cell-derived exosomes: a message in a bottle. Biochim Biophys Acta, 2012. 1826 (1): p. 103-11.
[8] Palazzo, A. F. and E. S. Lee, Non-coding RNA: what is functional and what is junk? Front Genet, 2015. 6: p. 2.
[9] Liu, Y., et al., The emerging role of the piRNA/piwi complex in cancer. Mol Cancer, 2019. 18 (1): p. 123.
[10] Bachellerie, J. P., J. Cavaille, and A. Huttenhofer, The expanding snoRNA world. Biochimie, 2002. 84 (8): p. 775-90.
[11] Jutzi, D., et al., Aberrant interaction of FUS with the U1 snRNA provides a molecular mechanism of FUS induced amyotrophic lateral sclerosis. Nat Commun, 2020. 11 (1): p. 6341.
[12] Long, Y., et al., How do lncRNAs regulate transcription? Sci Adv, 2017. 3 (9): p. eaao2110.
[13] Chan, J. J. and Y. Tay, Noncoding RNA: RNA Regulatory Networks in Cancer. Int J Mol Sci, 2018. 19 (5).
[14] Carpenter, S., et al., A long noncoding RNA mediates both activation and repression of immune response genes. Science, 2013. 341 (6147): p. 789-92.
[15] Luarte, A., et al., Potential Therapies by Stem Cell-Derived Exosomes in CNS Diseases: Focusing on the Neurogenic Niche. Stem Cells Int, 2016. 2016: p. 5736059.
[16] Lugli, G., et al., Plasma Exosomal miRNAs in Persons with and without Alzheimer Disease: Altered Expression and Prospects for Biomarkers. PLoS One, 2015. 10 (10): p. e0139233.
[17] Van Giau, V. and S. S. An, Emergence of exosomal miRNAs as a diagnostic biomarker for Alzheimer's disease. J Neurol Sci, 2016. 360: p. 141-52.
[18] Wickman, J. R., et al., Circulating microRNAs from the mouse tibia fracture model reflect the signature from patients with complex regional pain syndrome. Pain Rep, 2021. 6 (3): p. e950.
[19] Jean-Toussaint, R., et al., Therapeutic and prophylactic effects of macrophage-derived small extracellular vesicles in the attenuation of inflammatory pain. Brain Behav Immun, 2021. 94: p. 210-224.
[20] Hornick, N. I., et al., Serum Exosome MicroRNA as a Minimally-Invasive Early Biomarker of AML. Sci Rep, 2015. 5: p. 11295.
[21] Schwarzenbach, H., The clinical relevance of circulating, exosomal miRNAs as biomarkers for cancer. Expert Rev Mol Diagn, 2015. 15 (9): p. 1159-69.
[22] Johnson, L. R., et al., The immunostimulatory RNA RN7SL1 enables CAR-T cells to enhance autonomous and endogenous immune function. Cell, 2021. 184 (19): p. 4981-4995 e14.
[23] Pinho, A. G., et al., Cell Secretome: Basic Insights and Therapeutic Opportunities for CNS Disorders. Pharmaceuticals (Basel), 2020. 13 (2).
[24] Agrawal, G. K., et al., Plant secretome: unlocking secrets of the secreted proteins. Proteomics, 2010. 10 (4): p. 799-827.
[25] Baraniak, P. R. and T. C. McDevitt, Stem cell paracrine actions and tissue regeneration. Regen Med, 2010. 5 (1): p. 121-43.
[26] Hathout, Y., Approaches to the study of the cell secretome. Expert Rev Proteomics, 2007. 4 (2): p. 239-48.
[27] Tjalsma, H., et al., Signal peptide-dependent protein transport in Bacillus subtilis: a genome-based survey of the secretome. Microbiol Mol Biol Rev, 2000. 64 (3): p. 515-47.
[28] Vizoso, F. J., et al., Mesenchymal Stem Cell Secretome: Toward Cell-Free Therapeutic Strategies in Regenerative Medicine. Int J Mol Sci, 2017. 18 (9).
[29] Gnecchi, M., et al., Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med, 2005. 11 (4): p. 367-8.
[30] Teixeira, F. G., et al., Secretome of mesenchymal progenitors from the umbilical cord acts as modulator of neural/glial proliferation and differentiation. Stem Cell Rev Rep, 2015. 11 (2): p. 288-97.
[31] Brini, A. T., et al., Therapeutic effect of human adipose-derived stem cells and their secretome in experimental diabetic pain. Sci Rep, 2017. 7 (1): p. 9904.
[32] Khatab, S., et al., Mesenchymal stem cell secretome reduces pain and prevents cartilage damage in a murine osteoarthritis model. Eur Cell Mater, 2018. 36: p. 218-230.
[33] Gama, K. B., et al., Conditioned Medium of Bone Marrow-Derived Mesenchymal Stromal Cells as a Therapeutic Approach to Neuropathic Pain: A Preclinical Evaluation. Stem Cells Int, 2018. 2018: p. 8179013.
[34] Diomede, F., et al., Functional Relationship between Osteogenesis and Angiogenesis in Tissue Regeneration. Int J Mol Sci, 2020. 21 (9).
[35] Lee, S. and L. E. Goldfinger, RLIP76 regulates HIF-1 activity, VEGF expression and secretion in tumor cells, and secretome transactivation of endothelial cells. FASEB J, 2014. 28 (9): p. 4158-68.
[36] Asthana, A., et al., Secretome-Based Prediction of Three-Dimensional Hepatic Microtissue Physiological Relevance. ACS Biomater Sci Eng, 2020. 6 (1): p. 587-596.
[37] Rong, X., et al., Human fetal skin-derived stem cell secretome enhances radiation-induced skin injury therapeutic effects by promoting angiogenesis. Stem Cell Res Ther, 2019. 10 (1): p. 383.
[38] Salgado, A. J., et al., Adipose tissue derived stem cells secretome: soluble factors and their roles in regenerative medicine. Curr Stem Cell Res Ther, 2010. 5 (2): p. 103-10.
[39] Khaibullina, A. A., J. M. Rosenstein, and J. M. Krum, Vascular endothelial growth factor promotes neurite maturation in primary CNS neuronal cultures. Brain Res Dev Brain Res, 2004. 148 (1): p. 59-68.
[40] Guaiquil, V. H., et al., VEGF-B selectively regenerates injured peripheral neurons and restores sensory and trophic functions. Proc Natl Acad Sci U S A, 2014. 111 (48): p. 17272-7.
[41] Sosanya, N. M., et al., Involvement of brain-derived neurotrophic factor (BDNF) in chronic intermittent stress-induced enhanced mechanical allodynia in a rat model of burn pain. BMC Neurosci, 2019. 20 (1): p. 17.
[42] Eaton, S. L., et al., Total protein analysis as a reliable loading control for quantitative fluorescent Western blotting. PLoS One, 2013. 8 (8): p. e72457.
[43] Fontanilla, C. V., et al., Adipose-derived Stem Cell Conditioned Media Extends Survival time of a mouse model of Amyotrophic Lateral Sclerosis. Sci Rep, 2015. 5: p. 16953.
[44] Prochazka, V., et al., Therapeutic Potential of Adipose-Derived Therapeutic Factor Concentrate for Treating Critical Limb Ischemia. Cell Transplant, 2016. 25 (9): p. 1623-1633.
[45] Wei, X., et al., IFATS collection: The conditioned media of adipose stromal cells protect against hypoxia-ischemia-induced brain damage in neonatal rats. Stem Cells, 2009. 27 (2): p. 478-88.
[46] Wei, X., et al., Adipose stromal cells-secreted neuroprotective media against neuronal apoptosis. Neurosci Lett, 2009. 462 (1): p. 76-9.
[47] Zhao, L., et al., Adipose stromal cells-conditional medium protected glutamate-induced CGNs neuronal death by BDNF. Neurosci Lett, 2009. 452 (3): p. 238-40.
[48] Sweeney, B. A., et al., Exploring Non-Coding RNAs in RNAcentral. Curr Protoc Bioinformatics, 2020. 71 (1): p. e104.
[49] Siprashvili, Z., et al., The noncoding RNAs SNORD50A and SNORD50B bind K-Ras and are recurrently deleted in human cancer. Nat Genet, 2016. 48 (1): p. 53-8.
[50] Braicu, C., et al., The Function of Non-Coding RNAs in Lung Cancer Tumorigenesis. Cancers (Basel), 2019. 11 (5).
[51] Mannoor, K., et al., Small nucleolar RNA signatures of lung tumor-initiating cells. Mol Cancer, 2014. 13: p. 104.
[52] Liu, J., et al., Identification of potential prognostic small nucleolar RNA biomarkers for predicting overall survival in patients with sarcoma. Cancer Med, 2020. 9 (19): p. 7018-7033.
[53] Toms, D., et al., Small RNA sequencing reveals distinct nuclear microRNAs in pig granulosa cells during ovarian follicle growth. J Ovarian Res, 2021. 14 (1): p. 54.
[54] Fitz, N. F., et al., Small nucleolar RNAs in plasma extracellular vesicles and their discriminatory power as diagnostic biomarkers of Alzheimer's disease. Neurobiol Dis, 2021. 159: p. 105481.
[55] Lee, E. J., et al., Identification of piRNAs in the central nervous system. RNA, 2011. 17 (6): p. 1090-9.
[56] Phay, M., H. H. Kim, and S. Yoo, Analysis of piRNA-Like Small Non-coding RNAs Present in Axons of Adult Sensory Neurons. Mol Neurobiol, 2018. 55 (1): p. 483-494.
[57] Plestilova, L., et al., Expression and Regulation of PIWIL-Proteins and PIWI-Interacting RNAs in Rheumatoid Arthritis. PLoS One, 2016. 11 (11): p. e0166920.
[58] Zhang, C., et al., PiRNA-DQ541777 Contributes to Neuropathic Pain via Targeting Cdk5rap1. J Neurosci, 2019. 39 (45): p. 9028-9039.
[59] Spinelli, C., et al., Extracellular Vesicles as Conduits of Non-Coding RNA Emission and Intercellular Transfer in Brain Tumors. Noncoding RNA, 2018. 5 (1).
[60] Zhou, J., Y. Fan, and H. Chen, Analyses of long non-coding RNA and mRNA profiles in the spinal cord of rats using RNA sequencing during the progression of neuropathic pain in an SNI model. RNA Biol, 2017. 14 (12): p. 1810-1826.
[61] Mao, P., et al., Transcriptomic differential lncRNA expression is involved in neuropathic pain in rat dorsal root ganglion after spared sciatic nerve injury. Braz J Med Biol Res, 2018. 51 (10): p. e7113.
[62] Baskozos, G., et al., Comprehensive analysis of long noncoding RNA expression in dorsal root ganglion reveals cell-type specificity and dysregulation after nerve injury. Pain, 2019. 160 (2): p. 463-485.
[63] Jiang, B. C., et al., Identification of lncRNA expression profile in the spinal cord of mice following spinal nerve ligation-induced neuropathic pain. Mol Pain, 2015. 11: p. 43.
[64] Zhou, J., et al., Identification of the Spinal Expression Profile of Non-coding RNAs Involved in Neuropathic Pain Following Spared Nerve Injury by Sequence Analysis. Front Mol Neurosci, 2017. 10: p. 91.
[65] Yu, W., et al., LncRNA NONRATT021972 Was Associated with Neuropathic Pain Scoring in Patients with Type 2 Diabetes. Behav Neurol, 2017. 2017: p. 2941297.
[66] Yue, T., et al., Comprehensive analyses of long non-coding RNA expression profiles by RNA sequencing and exploration of their potency as biomarkers in psoriatic arthritis patients. BMC Immunol, 2019. 20 (1): p. 28.
[67] Mork, S., et al., Protein-driven inference of miRNA-disease associations. Bioinformatics, 2014. 30 (3): p. 392-7.
[68] Huang, L., F. Y. Deng, and S. F. Lei, Global correlation analysis for miRNA and protein expression profiles in human peripheral blood mononuclear cells. Mol Biol Rep, 2020. 47 (7): p. 5295-5304.
[69] Alshalalfa, M., miRNA regulation in the context of functional protein networks: principles and applications. Wiley Interdiscip Rev Syst Biol Med, 2014. 6 (2): p. 189-99.
[70] Pizzicannella, J., et al., Engineered Extracellular Vesicles From Human Periodontal-Ligament Stem Cells Increase VEGF/VEGFR2 Expression During Bone Regeneration. Front Physiol, 2019. 10: p. 512.
[71] Jin, Q., et al., Extracellular vesicles derived from human dental pulp stem cells promote osteogenesis of adipose-derived stem cells via the MAPK pathway. J Tissue Eng, 2020. 11: p. 2041731420975569.
[72] Ma, Y., et al., Induced neural progenitor cells abundantly secrete extracellular vesicles and promote the proliferation of neural progenitors via extracellular signal-regulated kinase pathways. Neurobiol Dis, 2019. 124: p. 322-334.
[73] Zhou, D., W. Zhai, and M. Zhang, Mesenchymal stem cell-derived extracellular vesicles promote apoptosis in RSC96 Schwann cells through the activation of the ERK pathway. Int J Clin Exp Pathol, 2018. 11 (11): p. 5157-5170.
[74] Song, X. S., et al., Activation of ERK/CREB pathway in spinal cord contributes to chronic constrictive injury-induced neuropathic pain in rats. Acta Pharmacol Sin, 2005. 26 (7): p. 789-98.
[75] Joo, J. D., et al., Lidocaine suppresses the increased extracellular signal-regulated kinase/cyclic AMP response element-binding protein pathway and pro-inflammatory cytokines in a neuropathic pain model of rats. Eur J Anaesthesiol, 2011. 28 (2): p. 106-11.
[76] Guo, J. R., et al., Effect and mechanism of inhibition of PI3K/Akt/mTOR signal pathway on chronic neuropathic pain and spinal microglia in a rat model of chronic constriction injury. Oncotarget, 2017. 8 (32): p. 52923-52934.
[77] Kiguchi, N., et al., Vascular endothelial growth factor signaling in injured nerves underlies peripheral sensitization in neuropathic pain. J Neurochem, 2014. 129 (1): p. 169-78.
[78] Kim, Y., et al., Amitriptyline inhibits the MAPK/ERK and CREB pathways and proinflammatory cytokines through A3AR activation in rat neuropathic pain models. Korean J Anesthesiol, 2019. 72 (1): p. 60-67.
[79] Lin, J. P., et al., Dexmedetomidine Attenuates Neuropathic Pain by Inhibiting P2X7R Expression and ERK Phosphorylation in Rats. Exp Neurobiol, 2018. 27 (4): p. 267-276.
[80] Ji, R. R., et al., MAP kinase and pain. Brain Res Rev, 2009. 60 (1): p. 135-48.
[81] Xu, X., et al., Microglial BDNF, PI3K, and p-ERK in the Spinal Cord Are Suppressed by Pulsed Radiofrequency on Dorsal Root Ganglion to Ease SNI-Induced Neuropathic Pain in Rats. Pain Res Manag, 2019. 2019: p. 5948686.
[82] Wang, B., et al., PKM2 is involved in neuropathic pain by regulating ERK and STAT3 activation in rat spinal cord. J Headache Pain, 2018. 19 (1): p. 7.
[83] Choi, S., et al., mTOR signaling intervention by Torin1 and XL388 in the insular cortex alleviates neuropathic pain. Neurosci Lett, 2020. 718: p. 134742.
[84] Tateda, S., et al., Rapamycin suppresses microglial activation and reduces the development of neuropathic pain after spinal cord injury. J Orthop Res, 2017. 35 (1): p. 93-103.
[85] Lopez-Bellido, R., et al., Growth Factor Signaling Regulates Mechanical Nociception in Flies and Vertebrates. J Neurosci, 2019. 39 (30): p. 6012-6030.
[86] Beazley-Long, N., et al., VEGFR2 promotes central endothelial activation and the spread of pain in inflammatory arthritis. Brain Behav Immun, 2018. 74: p. 49-67.
[87] Cho, Y. R., et al., Broussonetia kazinoki modulates the expression of VEGFR-2 and MMP-2 through the inhibition of ERK, Akt and p70S6K dependent signaling pathways: Its implication in endothelial cell proliferation, migration and tubular formation. Oncol Rep, 2014. 32 (4): p. 1531-6.
[88] Saryeddine, L., et al., EGF-Induced VEGF Exerts a PI3K-Dependent Positive Feedback on ERK and AKT through VEGFR2 in Hematological In Vitro Models. PLoS One, 2016. 11 (11): p. e0165876.
[89] Wang, W., et al., MicroRNA-497 suppresses angiogenesis by targeting vascular endothelial growth factor A through the PI3K/AKT and MAPK/ERK pathways in ovarian cancer. Oncol Rep, 2014. 32 (5): p. 2127-33.
[90] Pengcheng, S., et al., MicroRNA-497 suppresses renal cell carcinoma by targeting VEGFR-2 in ACHN cells. Biosci Rep, 2017. 37 (3).
[91] Zachary, I., Neuroprotective role of vascular endothelial growth factor: signalling mechanisms, biological function, and therapeutic potential. Neurosignals, 2005. 14 (5): p. 207-21.
[92] Ogunshola, O. O., et al., Paracrine and autocrine functions of neuronal vascular endothelial growth factor (VEGF) in the central nervous system. J Biol Chem, 2002. 277 (13): p. 11410-5.
[93] Tovar, Y. R. L. B. and R. Tapia, VEGF protects spinal motor neurons against chronic excitotoxic degeneration in vivo by activation of PI3-K pathway and inhibition of p38MAPK. J Neurochem, 2010. 115 (5): p. 1090-101.
[94] Swendeman, S., et al., VEGF-A stimulates ADAM17-dependent shedding of VEGFR2 and crosstalk between VEGFR2 and ERK signaling. Circ Res, 2008. 103 (9): p. 916-8.
[95] Ferreira, J. R., et al., IL-1beta-pre-conditioned mesenchymal stem/stromal cells' secretome modulates the inflammatory response and aggrecan deposition in intervertebral disc. Eur Cell Mater, 2021. 41: p. 431-453.
[96] Miranda, J. P., et al., The Secretome Derived From 3D-Cultured Umbilical Cord Tissue MSCs Counteracts Manifestations Typifying Rheumatoid Arthritis. Front Immunol, 2019. 10: p. 18.
[97] Dabrowska, S., et al., Immunomodulatory and Regenerative Effects of Mesenchymal Stem Cells and Extracellular Vesicles: Therapeutic Outlook for Inflammatory and Degenerative Diseases. Front Immunol, 2020. 11: p. 591065.
[98] Ghafouri-Fard, S., et al., The Emerging Role of Exosomes in the Treatment of Human Disorders With a Special Focus on Mesenchymal Stem Cells-Derived Exosomes. Front Cell Dev Biol, 2021. 9: p. 653296.
Cite This Article
  • APA Style

    Megan Rose Hershfield, Misty Marie Strain, Roger Chavez, Michaela Rae Priess, Alberto Mares, et al. (2022). Exploring the Secretome’s Biomarker and Analgesic Potential. American Journal of Psychiatry and Neuroscience, 10(2), 63-76. https://doi.org/10.11648/j.ajpn.20221002.12

    Copy | Download

    ACS Style

    Megan Rose Hershfield; Misty Marie Strain; Roger Chavez; Michaela Rae Priess; Alberto Mares, et al. Exploring the Secretome’s Biomarker and Analgesic Potential. Am. J. Psychiatry Neurosci. 2022, 10(2), 63-76. doi: 10.11648/j.ajpn.20221002.12

    Copy | Download

    AMA Style

    Megan Rose Hershfield, Misty Marie Strain, Roger Chavez, Michaela Rae Priess, Alberto Mares, et al. Exploring the Secretome’s Biomarker and Analgesic Potential. Am J Psychiatry Neurosci. 2022;10(2):63-76. doi: 10.11648/j.ajpn.20221002.12

    Copy | Download

  • @article{10.11648/j.ajpn.20221002.12,
      author = {Megan Rose Hershfield and Misty Marie Strain and Roger Chavez and Michaela Rae Priess and Alberto Mares and Aarti Gautam and George Dimitrov and Ruoting Yang and Alex Valdez Trevino and Col Kenney Wells and Col Thomas Stark and Rasha Hammamieh and John Leo Clifford and Natasha Marie Sosanya},
      title = {Exploring the Secretome’s Biomarker and Analgesic Potential},
      journal = {American Journal of Psychiatry and Neuroscience},
      volume = {10},
      number = {2},
      pages = {63-76},
      doi = {10.11648/j.ajpn.20221002.12},
      url = {https://doi.org/10.11648/j.ajpn.20221002.12},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.ajpn.20221002.12},
      abstract = {Diagnostic and prognostic biomarkers of nerve injury and/or pain as well as improved pain therapeutics are needed, both on the battlefield to treat injured Service Members and in the civilian sector. Our previous research indicates that there are several differentially expressed (DE) extracellular vesicle-derived microRNAs (EV-miRNAs) isolated from rat plasma following spinal nerve ligation (SNL). As such, EV-miRNAs hold promise as biomarkers and therapeutic targets. The secretome contains biological mediators, including EVs, which are released into the extracellular space. In this study we focus on evaluating EV-non-coding RNAs (ncRNAs), examine effects of SNL on key protein expression in the prefrontal cortex (PFC), and test the secretome’s analgesic properties. To accomplish these goals, anesthetized male Sprague Dawley rats underwent SNL and nociceptive behavior measurements, plasma collection followed by EV RNA isolation, small RNA sequencing, and analysis. Expression of several key proteins in the PFC was determined by Wes/Jess analysis. The secretome bath was applied directly to the ligated nerve and the paw withdrawal threshold (PWT) was measured. We identified differences in several classes of ncRNAs such as piRNAs, snoRNAs, and snRNAs post-SNL. Levels of phosphorylated forms of P70S6K and ERK1 were increased in the dorsal PFC at 15 days post-SNL. Bath application of the secretome directly to the ligated nerve resulted in recovery of the reduced PWT (increased mechanical sensitivity) that is induced by SNL. Here, we have identified specific EV-ncRNAs that could contribute to the formation of pain. Furthermore, we have evaluated a novel product for analgesic efficacy that could function to exploit the underlying mechanisms that contribute to pain development, thus reducing acute pain. This is key in treating Service Members on the battlefield in order to prevent pain chronification.},
     year = {2022}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - Exploring the Secretome’s Biomarker and Analgesic Potential
    AU  - Megan Rose Hershfield
    AU  - Misty Marie Strain
    AU  - Roger Chavez
    AU  - Michaela Rae Priess
    AU  - Alberto Mares
    AU  - Aarti Gautam
    AU  - George Dimitrov
    AU  - Ruoting Yang
    AU  - Alex Valdez Trevino
    AU  - Col Kenney Wells
    AU  - Col Thomas Stark
    AU  - Rasha Hammamieh
    AU  - John Leo Clifford
    AU  - Natasha Marie Sosanya
    Y1  - 2022/05/10
    PY  - 2022
    N1  - https://doi.org/10.11648/j.ajpn.20221002.12
    DO  - 10.11648/j.ajpn.20221002.12
    T2  - American Journal of Psychiatry and Neuroscience
    JF  - American Journal of Psychiatry and Neuroscience
    JO  - American Journal of Psychiatry and Neuroscience
    SP  - 63
    EP  - 76
    PB  - Science Publishing Group
    SN  - 2330-426X
    UR  - https://doi.org/10.11648/j.ajpn.20221002.12
    AB  - Diagnostic and prognostic biomarkers of nerve injury and/or pain as well as improved pain therapeutics are needed, both on the battlefield to treat injured Service Members and in the civilian sector. Our previous research indicates that there are several differentially expressed (DE) extracellular vesicle-derived microRNAs (EV-miRNAs) isolated from rat plasma following spinal nerve ligation (SNL). As such, EV-miRNAs hold promise as biomarkers and therapeutic targets. The secretome contains biological mediators, including EVs, which are released into the extracellular space. In this study we focus on evaluating EV-non-coding RNAs (ncRNAs), examine effects of SNL on key protein expression in the prefrontal cortex (PFC), and test the secretome’s analgesic properties. To accomplish these goals, anesthetized male Sprague Dawley rats underwent SNL and nociceptive behavior measurements, plasma collection followed by EV RNA isolation, small RNA sequencing, and analysis. Expression of several key proteins in the PFC was determined by Wes/Jess analysis. The secretome bath was applied directly to the ligated nerve and the paw withdrawal threshold (PWT) was measured. We identified differences in several classes of ncRNAs such as piRNAs, snoRNAs, and snRNAs post-SNL. Levels of phosphorylated forms of P70S6K and ERK1 were increased in the dorsal PFC at 15 days post-SNL. Bath application of the secretome directly to the ligated nerve resulted in recovery of the reduced PWT (increased mechanical sensitivity) that is induced by SNL. Here, we have identified specific EV-ncRNAs that could contribute to the formation of pain. Furthermore, we have evaluated a novel product for analgesic efficacy that could function to exploit the underlying mechanisms that contribute to pain development, thus reducing acute pain. This is key in treating Service Members on the battlefield in order to prevent pain chronification.
    VL  - 10
    IS  - 2
    ER  - 

    Copy | Download

Author Information
  • Pain and Sensory Trauma, US Army Institute of Surgical Research, JBSA Fort Sam Houston, USA

  • Pain and Sensory Trauma, US Army Institute of Surgical Research, JBSA Fort Sam Houston, USA

  • Pain and Sensory Trauma, US Army Institute of Surgical Research, JBSA Fort Sam Houston, USA

  • Pain and Sensory Trauma, US Army Institute of Surgical Research, JBSA Fort Sam Houston, USA

  • Pain and Sensory Trauma, US Army Institute of Surgical Research, JBSA Fort Sam Houston, USA

  • Medical Readiness Systems Biology Branch, Walter Reed Army Institute of Research, Silver Spring, USA

  • The Geneva Foundation, Tacoma, USA

  • Medical Readiness Systems Biology Branch, Walter Reed Army Institute of Research, Silver Spring, USA

  • Pain and Sensory Trauma, US Army Institute of Surgical Research, JBSA Fort Sam Houston, USA

  • Pain and Sensory Trauma, US Army Institute of Surgical Research, JBSA Fort Sam Houston, USA

  • Pain and Sensory Trauma, US Army Institute of Surgical Research, JBSA Fort Sam Houston, USA

  • Medical Readiness Systems Biology Branch, Walter Reed Army Institute of Research, Silver Spring, USA

  • Pain and Sensory Trauma, US Army Institute of Surgical Research, JBSA Fort Sam Houston, USA

  • Pain and Sensory Trauma, US Army Institute of Surgical Research, JBSA Fort Sam Houston, USA

  • Sections