Acta Scientific Neurology (ASNE) (ISSN: 2582-1121)

Review Article Volume 5 Issue 12

BDNF: The Old-New Pain Mediator and Modulator of Neuropathic Pain and Neuroinflammation

Bilal F Shanti*, Zaynab I Shanti and Ihsan F Shanti

Department of Pain Medicine, Omnia Pain Consultants, USA

*Corresponding Author: Bilal F Shanti, Department of Pain Medicine, Omnia Pain Consultants, USA.

Received: September 10, 2022; Published: November 08, 2022

Abstract

This manuscript will review the crucial role of BDNF as a pain mediator and modulator of neuroinflammation and neuropathic pain. BDNF belongs to of the neurotrophin family of growth factors that is encoded by the bdnf gene. Its role as modulator was found to act on neuronal excitability and synaptic plasticity has triggered huge interest in neurobiological pain research. This pointed to renewed interest as a potential target for pharmacological agents in its long cascade of mechanism of actions.

There is no clear-cut explanation on how BDNF works. One thing we know is that BDNF mechanism of action depends on whether we are dealing with a normal nerve, after an acute injury, or following chronic state. It is considered a major tertiary mediator in the genesis of central sensitization. It mediates inflammatory and peripheral injury-induced pain and play a role by increasing dorsal horn excitability. Hence disrupting these cycles can potentially curb the genesis of chronic pain.

Some suggested mechanisms of action of BDNF in neuropathic pain is via enhanced neuronal sensitivity to painful stimuli and an enhanced co-expression of thermo-TRP channels. Another possible mechanism entails the involvement of microglia in pain. Following a peripheral injury, GABA-BDNF plays an essential role in formation of neuropathic pain. BDNF increases both the excitatory and inhibitory synaptic drives to putative excitatory interneurons while attenuating synaptic transmission to inhibitory GABAergic neurons.

In the absence of any injury and in normal states, BDNF-mediated effects contribute to pain. An increase in BDNF and TrkB expression activate MAPK/ERK and PLC-PKC kinase pathways.

In the acute stage of nerve injury, the levels of both BDNF and TrkB are decreased in the spinal cord dorsal horn. During the chronic stage of injury, pERK levels are also noted to increase. This may result from increases in both BDNF-TrkB signaling and TNFš›¼-TNFR signaling.

BDNF may also potentially modulate spinal neuron responsiveness by potentiation of postsynaptic N-methyl-D-aspartate (NMDA) receptors.

BDNF noticeably enhanced the frequency of miniature excitatory postsynaptic currents (EPSCs) recorded in superficial dorsal horn neurons.

All these mechanisms and theories may potentially be a dream for clinicians to work on and modulate pain.

Keywords:BDNF; TrkB; Plasticity; Neuropathic Pain; Neurotrophins; Neuroinflammation; Plasticity; NMDA Receptors; IL-1Ī²; GABA; Central Sensitization

References

  1. Nijs J., et al. “Brainā€derived neurotrophic factor as a driving force behind neuroplasticity in neuropathic and central sensitization pain: a new therapeutic target?” Expert Opinion on Therapeutic Targets 4 (2015): 565-576.
  2. Pezet S and McMahon SB. “Neurotrophins: mediators and modulators of pain”. Annual Review of Neuroscience 29 (2006): 507-538.
  3. Keefe KM., et al. “Targeting Neurotrophins to Specific Populations of Neurons: NGF, BDNF, and NT-3 and Their Relevance for Treatment of Spinal Cord Injury”. International Journal of Molecular Sciences 3 (2017): 548.
  4. Merighi A., et al. “BDNF as a pain modulator”. Progress in Neurobiology85.3 (2008): 297-317.
  5. Yamamoto H and Gurney ME. “Human platelets contain brain-derived neurotrophic factor”. Journal of Neuroscience 10.11 (1990): 3469-3478.
  6. Miranda M., et al. “Brainā€derived neurotrophic factor: a key molecule for memory in the healthy and the pathological brain”. Frontiers in Cellular Neuroscience 13 (2019):
  7. Garraway SM and Huie JR. “Spinal plasticity and behavior: BDNF-induced neuromodulation in uninjured and injured spinal cord”. Neural Plasticity (2016).
  8. Levine ES., et al. “Brain-derived neurotrophic factor rapidly enhances synaptic transmission in hippocampal neurons via postsynaptic tyrosine kinase receptors”. Proceedings of the National Academy of Sciences of the United States of America17 (1995): 8074-8077.
  9. Gottmann K., et al. “BDNF signaling in the formation, maturation and plasticity of glutamatergic and GABAergic synapses”. Experimental Brain Research3-4 (2009): 203-234.
  10. Halievski K., et al. “Sex-dependent mechanisms of chronic pain: a focus on microglia and P2X4R”. Journal of Pharmacology and Experimental Therapeutics 375 (2020): 202-209.
  11. Boakye PA., et al. “Mediators of Neuropathic Pain; Focus on Spinal Microglia, CSF-1, BDNF, CCL21, TNF-α, Wnt Ligands, and Interleukin 1β”. Frontiers in Pain Research (Lausanne) 2 (2021):
  12. Alles SRA and Smith PA. “The etiology and pharmacology of neuropathic pain”. Pharmacological Reviews 70 (2018): 315-347.
  13. Guan Z., et al. “Injured sensory neuron-derived CSF1 induces microglial proliferation and DAP12- dependent pain”. Nature Neuroscience 19 (2016): 94-101.
  14. Ulmann L., et al. “Up-regulation of P2X4 receptors in spinal microglia after peripheral nerve injury mediates BDNF release and neuropathic pain”. Journal of Neuroscience 28 (2008): 11263-11268.
  15. Mapplebeck JCS., et al. “Microglial P2X4R-evoked pain hypersensitivity is sexually dimorphic in rats”. Pain 159 (2018): 1752-1763.
  16. Zhang X and Li G. “P2Y receptors in neuropathic pain”. Pharmacology Biochemistry and Behavior 186 (2019):
  17. Zhang W., et al. “Neuron activity-induced Wnt signaling up-regulates expression of brain-derived neurotrophic factor in the pain neural circuit”. Journal of Biological Chemistry 293 (2018): 15641-15651.
  18. Ciobanu C., et al. “Acute and chronic effects of neurotrophic factors BDNF and GDNF on responses mediated by thermo-sensitive TRP channels in cultured rat dorsal root ganglion neurons”. Brain Research 1284 (2009): 54-67.
  19. Ulmann L., et al. “Up-regulation of P2X4 receptors in spinal microglia after peripheral nerve injury mediates BDNF release and neuropathic pain”. Journal of Neuroscience 44 (2008): 11263-11268.
  20. Garraway SM., et al. “Intermittent noxious stimulation following spinal cord contusion injury impairs locomotor recovery and reduces spinal brain-derived neurotrophic factor-tropomyosin-receptor kinase signaling in adult rats”. Neuroscience 199 (2011): 86-102.
  21. Garraway SM and Huie JR. “Spinal plasticity and behavior: BDNF-induced neuromodulation in uninjured and injured spinal cord”. Neural Plasticity (2016).
  22. Garraway SM., et al. “Peripheral noxious stimulation reduces withdrawal threshold to mechanical stimuli after spinal cord injury: role of tumor necrosis factor alpha and apoptosis”. Pain11 (2014): 2344-2359.
  23. South SM., et al. “A conditional deletion of the NR1 subunit of the NMDA receptor in adult spinal cord dorsal horn reduces NMDA currents and injury-induced pain”. Journal of Neuroscience 12 (2003): 5031-5040.
  24. Garraway SM., et al. “BDNF sensitizes the response of lamina II neurons to high threshold primary afferent inputs”. European Journal of Neuroscience 9 (2003): 2467-2476.
  25. Slack SE., et al. “Brain-derived neurotrophic factor induces NMDA receptor subunit one phosphorylation via ERK and PKC in the rat spinal cord”. European Journal of Neuroscience 7 (2004): 1769-1778.
  26. Matayoshi S., et al. “Actions of brain-derived neurotrophic factor on spinal nociceptive transmission during inflammation in the rat”. The Journal of Physiology Pt.2 (2005): 685-695.
  27. Coull JA., et al. “BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain”. Nature 7070 (2005): 1017-1021.
  28. Pezet S., et al. “BDNF modulates sensory neuron synaptic activity by a facilitation of GABA transmission in the dorsal horn”. Molecular and Cellular Neuroscience 1 (2002a): 51-62.
  29. Lever I., et al. “Release of BDNF and GABA in the dorsal horn of neuropathic rats”. European Journal of Neuroscience 5 (2003): 1169-1174.
  30. Siuciak JA., et al. “Antinociceptive effect of brain-derived neurotrophic factor and neurotrophin-3”. Brain Research1-2 (1994): 326-330.
  31. Shih-Jen Tsai. “Possible involvement of brain-derived neurotrophic factor in the antinociceptive effect of antidepressants in neuropathic pain”. Medical Hypotheses3 (2005): 530-533.
  32. Bardoni R., et al. “BDNF-mediated modulation of GABA and glycine release in dorsal horn lamina II from postnatal rats”. Developmental Neurobiology 7 (2007): 960-975.
  33. Huang YJ., et al. “Complete spinal cord injury (SCI) transforms how brain derived neurotrophic factor (BDNF) affects nociceptive sensitization”. Experimental Neurology 288 (2017): 38-50.
  34. Klein K., et al. “ATP induced brain-derived neurotrophic factor expression and release from osteoarthritis synovial fibroblasts is mediated by purinergic receptor P2X4”. PLoS One7.5 (2012):
  35. Groth R and Aanonsen L. “Spinal brain-derived neurotrophic factor (BDNF) produces hyperalgesia in normal mice while antisense directed against either BDNF or trkB, prevent inflammation-induced hyperalgesia”. Pain100.1-2 (2002): 171-181.
  36. Melemedjian OK., et al. “BDNF regulates atypical PKC at spinal synapses to initiate and maintain a centralized chronic pain state”. Molecular Pain 9 (2013):
  37. Kerr BJ., et al. “Brain-derived neurotrophic factor modulates nociceptive sensory inputs and NMDA-evoked responses in the rat spinal cord”. Journal of Neuroscience 12 (1999): 5138-5148.
  38. Bao Y., et al. “PAR2-mediated upregulation of BDNF contributes to central sensitization in bone cancer pain”. Molecular Pain10 (2014): 1744-8069.
  39. Hökfelt T., et al. “Messenger plasticity in primary sensory neurons following axotomy and its functional implications”. Trends in Neurosciences 1 (1994): 22-30.
  40. Lalisse S., et al. “Sensory neuronal P2RX4 receptors controls BDNF signaling in inflammatory pain”. Scientific Reports 8.1 (2018), 1-12.
  41. Cho HJ., et al. “Increased brain-derived neurotrophic factor immunoreactivity in rat dorsal root ganglia and spinal cord following peripheral inflammation”. Brain Research1-2 (1997): 269-272.
  42. Mannion RJ., et al. “Neurotrophins: peripherally and centrally acting modulators of tactile stimulus-induced inflammatory pain hypersensitivity”. Proceedings of the National Academy of Sciences of the United States of America 16 (1999): 9385-9390.
  43. Cao T., et al. “Function and mechanisms of truncated BDNF receptor TrkB. T1 in neuropathic pain”. Cells 9.5 (2020): 1194.
  44. Marcol W., et al. “BDNF contributes to animal model neuropathic pain after peripheral nerve transection”. Neurosurgical Review 30 (2007): 235-243.
  45. Fernyhough P., et al. “Altered neurotrophin mRNA levels in peripheral nerve and skeletal muscle of experimentally diabetic rats”. Journal of Neurochemistry 64 (1995): 1231-1237.
  46. Cao XH., et al. “Reduction in voltage-gated K (+)channel activity in primary sensory neurons in painful diabetic neuropathy: Role of brain-derived neurotrophic factor”. Journal of Neurochemistry 114 (2010): 1460-1475.
  47. Khan N and Smith MT. “Neurotrophins and neuropathic pain: role in pathobiology”. Molecules 20.6 (2015): 10657-10688.
  48. Sobue G., et al. “Expression of mRNAs for neurotrophins (NGF, BDNF and NT-3) and their receptors (p75NGFR, Trk, TrkB, and TrkC) in human peripheral neuropathies”. Neurochemical Research 23 (1998): 821-829.
  49. Azoulay D and Horowitz NA. “Brain-derived neurotrophic factor as a potential biomarker of chemotherapy-induced peripheral neuropathy and prognosis in hematological malignancies; what we have learned, the challenges and a need for global standardization”. British Journal of Haematology 191 (2020): 17-18.
  50. Apfel SC and Kessler JA. “Neurotrophic factors in the therapy of peripheral neuropathy”. Bailliere's Clinical Neurology 4 (1995): 593-606.
  51. Shanti BF., et al. “Metamorphosis of Acute to Chronic Pain: A Novel Era of Discoveries in the Field of Pain Medicine". Acta Scientific Neurology 12 (2021): 07-11.
  52. Ling DS., et al. “Protein kinase Mzeta is necessary and sufficient for LTP maintenance”. Nature Neuroscience 5 (2002): 295-296.
  53. Jo Nijs., et al. “Brain-derived neurotrophic factor as a driving force behind neuroplasticity in neuropathic and central sensitization pain: a new therapeutic target?”Expert Opinion on Therapeutic Targets4 (2015): 565-576.
  54. Sikandar S., et al. “Brain-derived neurotrophic factor derived from sensory neurons plays a critical role in chronic pain”. Brain4 (2018): 1028-1039.

Citation

Citation: Bilal F Shanti., et al. ā€œBDNF: The Old-New Pain Mediator and Modulator of Neuropathic Pain and Neuroinflammation". Acta Scientific Neurology 5.12 (2022): 33-42.

Copyright

Copyright: Ā© 2022 Bilal F Shanti., et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.




Metrics

Acceptance rate32%
Acceptance to publication20-30 days

Indexed In




News and Events


  • Certification for Review
    Acta Scientific certifies the Editors/reviewers for their review done towards the assigned articles of the respective journals.
  • Submission Timeline for Upcoming Issue
    The last date for submission of articles for regular Issues is April 30th, 2024.
  • Publication Certificate
    Authors will be issued a "Publication Certificate" as a mark of appreciation for publishing their work.
  • Best Article of the Issue
    The Editors will elect one Best Article after each issue release. The authors of this article will be provided with a certificate of "Best Article of the Issue".
  • Welcoming Article Submission
    Acta Scientific delightfully welcomes active researchers for submission of articles towards the upcoming issue of respective journals.

Contact US