Summary

Traumatic Peripheral Nerve Injury in Mice

Published: March 25, 2022
doi:

Summary

The present protocol describes traumatic peripheral nerve injuries (TPNIs), including precisely calibrated crush, strictly aligned and misaligned laceration, as well as grafted and non-grafted gaps of the sciatic nerve in mice. Custom-designed sensors are developed to gauge nerve trauma, induced with commonly available tools to ensure reproducible post-TPNI outcomes.

Abstract

Traumatic peripheral nerve injury (TPNI) is a common cause of morbidity following orthopedic trauma. Reproducible and precise methods of injuring nerve and denervating muscle have long been a goal in musculoskeletal research. Many traumatically injured limbs have nerve trauma that defines the long-term patient outcome. Over several years, precise methods of producing microsurgical nerve injuries have been developed, including crush, lacerations, and nerve-gap grafting, allowing for reproducible outcome assessments. Moreover, newer methods are created for calibrated crush injuries that offer clinically relevant correlations with outcomes used to assess human patients. The principles of minimal manipulation to ensure low variability in nerve injury allow for adding still more associated tissue injuries into these models. This includes direct muscle crush and other components of limb injury. Finally, atrophy assessment and precise analysis of behavioral outcomes make these methods a complete package for studying musculoskeletal trauma that realistically incorporates all the elements of human traumatic limb injury.

Introduction

Traumatic peripheral nerve injury (TPNI) is a common cause of morbidity after orthopedic trauma1,2,3. Yearly, approximately 3% of trauma patients suffer nerve injury1,4, at an incidence of 3,50,000 cases5, resulting in 50,000 surgical repairs6. TPNIs occur in a wide range of severity, and the functional recovery directly depends on the type and severity of these injuries7,8,9. Less severe trauma (e.g., mild crush, incomplete laceration, etc.) will injure the myelin sheath and axons first, while more severe forces (e.g., severe crush, complete lacerations, etc.) will disrupt the connective nerve tissues; for example, the endoneurium, perineurium, and epineurium in addition to the myelin and axons1,10. Patients with TPNI hope that nerve function will eventually return, and muscle atrophy will be reversed. Decades of research have provided no precise treatments to enhance or ensure complete recovery despite advances in the treatment procedures11,12.

Nerve transections will not heal without surgical repair, which is often performed under the microscope. Repairs are typically performed end-to-end, making an effort to ensure that the repair site is under no tension. Nerve grafting is used to ensure that repairs are tensionless13,14. Despite the seemingly advanced methods used in these repairs, functional recovery is generally unimpressive11,12. Rehabilitation is often incomplete and unsatisfying. The optimal functional recovery requires regenerating axons to cross the injury site (nerve bridge) and innervate the target organ. These processes are complicated by axonal misdirection or growth stunting, resulting in muscle atrophy and eventual failure to recover15,16,17,18. It has been shown that functional outcomes following nerve repair (e.g., end-to-end suturing, isografting, etc.) depend on the accuracy of fascicular apposition19,20. Proper directionality of the transected nerve stumps and their fascicles is thus critical in nerve repair, without which poor functional recovery can be expected even with optimal axonal regeneration. Microsurgical suture repair itself is a traumatic process, and little has occurred in terms of novel methods to improve outcomes drastically. The field lacks reproducible nerve transection animal models, which result in predictable gaps that allow reliable recovery measurements on a functional and tissue level. Such methods, if available, would allow characterization of nerve regeneration without the problems of variable changes in neural vascularization and post denervation atrophy21,22. Many groups endeavor to use better models that limit this kind of variability. One way is to ensure that nerve repairs are minimally manipulated, and nerve stumps are perfectly opposed.

This is best accomplished by using a standardized peripheral nerve transection technique called stepwise cut and fibrin glue (STG). Repairs in this STG model are secured with fibrin glue, and gap distances are standardized and minimized21,22. Fibrin glue itself is employed in humans for these repairs, likely for the same reasons, along with its beneficial effects on post-repair scar formation23,24. The key to the present method is that the nerve repair begins before the laceration is completed, ensuring a fixed injury pattern. This current method exhibited a close commonality to the characteristic pathophysiology of nerve transection with the gold-standard epineural suturing, and the negative impact of fibrin glue was not observed on nerve regeneration. Repairing of the sciatic nerve transection with fibrin glue in mice ameliorates the elongation of axon compared to early nerve regeneration via suturing, and these findings are consistent with STG. STG also benefits from the minimal manipulation principle, where the nerve is never touched for suture positioning21. This effectively standardizes the nerve trauma associated with repair in the model. Similar principles were used to investigate misalignment by flipping the nerve before gluing22. This allowed direct comparison of nerve injuries where almost the same amount of manipulation contributed to differences in alignment without increased gap or trauma. This facilitated the direct examination of the effect of alignment on nerve-injury-induced neurovascular changes21,22, muscle atrophy21,22, and functional recovery21,22. The present investigation is all that allows the study of purposefully and precisely misaligned nerve stumps.

Most of the nerves in TPNI are not severed, have no gap or defect, and seem to be capable of recovery, and yet in many of these cases, limbs remain permanently dysfunctional from nerve injuries and confound interventions. Experimental TPNIs are customarily performed on rodent sciatic nerve crush injuries (SNCIs) using locking needle drivers (NDs), forceps, or similar devices, and an experienced surgeon to create a precise and reproducible crush injury25,26,27,28,29,30. SNCI animal models depend on innate operator precision to limit pressure variation, but this is never measured explicitly. This results in variability between animals and studies, with no clear guidance on the standardized pressure. It is thus anticipated that the capability to precisely deliver and report a coherent, accurate series of injuries with various known intensities may benefit the TPNI field. A perfect model can provide an SNCI of a known nerve injury severity to each animal by any lab or researcher for authentic inter-study and device replicability. To address this deficiency, a unique calibrated digital device was constructed containing a Force Sensitive Resistor (FSR), proficient in reporting the pressure (real-time) applied to a nerve. This device was then tested for the replicability of various crush injury pressures deployed by diverse types of forceps and NDs31.

Finally, a specific method was developed to address gaps in nerve32. The nerve gaps in the literature are induced by removing a nerve section and then repairing it back into the defect13,33,34. The manipulation required for this surgical procedure is often compounded with suturing, and the stumps of the nerve retract variably21,32,34. It was based on the reasoning that using isogenic oversized nerve grafts, the nerve stump retraction will never an issue32. The method required the simultaneous operation on two or three animals at once, taking a 7 mm graft to place into a 5 mm defect induced in another animal. The defect size of the second animal was then used to graft a still smaller defect in another animal if needed. This resulted in a comprehensive method for simultaneous surgery to graft defects with donor nerves that are always larger than the nerve defect to ensure tensionless repair. On coupling with the requirement of minimal manipulation, this offers an avenue to study graft length directly in syngeneic animals without asymmetric graft gaps that are ubiquitous in the literature20,32,34.

Protocol

The experimental design and animal protocols were approved by the Institutional Animal Care and Use Committee (IACUC) at Penn State University College of Medicine. Adult C57BL/6J male mice, 10-week-old, weighing 20-25 g, were used for the studies. Animals were housed at the animal facility under sterile animal management conditions, and they were acclimatized at least 5 days before conducting the studies. 1. Animal preparation Anesthetize the animals deeply using a…

Representative Results

The custom-made digital pressure sensor device (Figure 1D) operates by detecting the change in resistance of the FSR when a force is applied. This device senses and records the most modest pressure amounts applied to it with a response time of <5 µs, a sampling rate of 20 Hz, and a pressure range of 2.5-25 lbs31. The differences in the forceps (Figure 1C) induced SNCI (Figure 1A,B) p…

Discussion

The history of TPNI research stretches over several decades11,12. Early experiments with dogs and larger species established the importance of animal models in the study of TPNI outcomes36,37,38. Over time, these models have moved into smaller rodents, with their established and commonly used validated outcomes measures39,<sup class="xr…

Disclosures

The authors have nothing to disclose.

Acknowledgements

This work was supported by grants from the NIH (K08 AR060164-01A) and DOD (W81XWH-16-1-0725; W81XWH-19-1-0773) in addition to institutional support from the Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.

Materials

Alcohol prep COVIDIEN 5110
Buprenorphine ZooPharm BSRLAB0.5-211706
C57BL/6J Jackson Laboratories, Bar Harbor N/A
Cotton tipped applicators Puritan 25-8062WC
Dissecting scissor ASSI ASSI.SDC18R8
Fibrin glue-TISSEEL Baxter 1501263
Force Sensitive Resistor (FSR) N/A FlexiForce A301
Forceps FST-Dumont 5SF Inox, 11252-00
GraphPad Prism GraphPad Software Inc. Version 8.4.3.
Homeothermic heating pad Kent Scientific RJ1675
Ketamine/Ketaved VEDCO VED1220
Microsurgical Forceps Miltex Premium instruments BL1901
Ophthalmic lubricant ointment Akorn Animal Health NDC 59399-162-35
Petri dish VWR 25384-092
Phosphate-buffered saline Gibco 14190-144
Povidone iodine Solimo L0017765SA
Precision pinch pressure sensor device Custom made N/A
Scissor Miltex Premium 21-536
Stereo zoom binocular microscope World Precision Instruments Model PZMIII
Sterile gloves Cardinal Health 9L19E511
Surgical staples 3M-Precise DS-25
Surgical Tape 3M-Microphore 1530-0
Sutures Ethicon BV130-5
Syringe BD syringe 309597
Trimmer Philips Electronics MG3750
Xylazine/Anased Akorn Animal Health, Inc. VAM4811

References

  1. Robinson, L. R. Traumatic injury to peripheral nerves. Muscle & Nerve. 23 (6), 863-873 (2000).
  2. Campbell, W. W. Evaluation and management of peripheral nerve injury. Clinical Neurophysiology. Official Journal of the International Federation of Clinical Neurophysiology. 119 (9), 1951-1965 (2008).
  3. Dirschl, D. R., Dahners, L. E. The mangled extremity: when should it be amputated. The Journal of the American Academy of Orthopaedic Surgeons. 4 (4), 182-190 (1996).
  4. Noble, J., Munro, C. A., Prasad, V. S., Midha, R. Analysis of upper and lower extremity peripheral nerve injuries in a population of patients with multiple injuries. The Journal of Trauma. 45 (1), 116-122 (1998).
  5. Barton, N., Kelsey, J., Paemer, A., Nelson, L., Felberg, A., Rice, D. Upper extremity disorders: frequency, impact and cost. Journal of Hand Surgery. 23 (2), 255 (1998).
  6. Evans, G. R. Peripheral nerve injury: a review and approach to tissue engineered constructs. The Anatomical Record. 263 (4), 396-404 (2001).
  7. Lundborg, G. . Nerve injury and repair: regeneration, reconstruction, and cortical remodeling. , (2004).
  8. Thieme, . Surgery of peripheral nerves: a case-based approach. , (2008).
  9. Brushart, T. M. . Nerve repair. , (2011).
  10. Menorca, R. M. G., Fussell, T. S., Elfar, J. C. Nerve physiology: mechanisms of injury and recovery. Hand Clinics. 29 (3), 317-330 (2013).
  11. Faroni, A., Mobasseri, S. A., Kingham, P. J., Reid, A. J. Peripheral nerve regeneration: experimental strategies and future perspectives. Advanced Drug Delivery Reviews. 82-83, 160-167 (2015).
  12. Zochodne, D. W. The challenges and beauty of peripheral nerve regrowth. Journal of the peripheral nervous system: JPNS. 17 (1), 1-18 (2012).
  13. Isaacs, J. Major peripheral nerve injuries. Hand Clinics. 29 (3), 371-382 (2013).
  14. Siemionow, M., Brzezicki, G. Chapter 8: Current techniques and concepts in peripheral nerve repair. International Review of Neurobiology. 87, 141-172 (2009).
  15. de Ruiter, G. C. W., et al. Misdirection of regenerating motor axons after nerve injury and repair in the rat sciatic nerve model. Experimental Neurology. 211 (2), 339-350 (2008).
  16. Gordon, T. Nerve regeneration: understanding biology and its influence on return of function after nerve transfers. Hand Clinics. 32 (2), 103-117 (2016).
  17. Sobotka, S., Mu, L. Muscle reinnervation with nerve-muscle-endplate band grafting technique: correlation between force recovery and axonal regeneration. The Journal of Surgical Research. 195 (1), 144-151 (2015).
  18. Witzel, C., Rohde, C., Brushart, T. M. Pathway sampling by regenerating peripheral axons. The Journal of Comparative Neurology. 485 (3), 183-190 (2005).
  19. Orgel, M. G., Terzis, J. K. Epineurial vs. perineurial repair. Plastic and Reconstructive Surgery. 60 (1), 80-91 (1977).
  20. Bassilios Habre, S., Bond, G., Jing, X. L., Kostopoulos, E., Wallace, R. D., Konofaos, P. The surgical management of nerve gaps: present and future. Annals of Plastic Surgery. 80 (3), 252-261 (2018).
  21. Lee, J. I., et al. A novel nerve transection and repair method in mice: histomorphometric analysis of nerves, blood vessels, and muscles with functional recovery. Scientific Reports. 10 (1), 21637 (2020).
  22. Lee, J. I., et al. Purposeful misalignment of severed nerve stumps in a standardized transection model reveals persistent functional deficit with aberrant neurofilament distribution. Military Medicine. 186, 696-703 (2021).
  23. Sameem, M., Wood, T. J., Bain, J. R. A systematic review on the use of fibrin glue for peripheral nerve repair. Plastic and Reconstructive Surgery. 127 (6), 2381-2390 (2011).
  24. Spotnitz, W. D. Fibrin sealant: the only approved hemostat, sealant, and adhesive-a laboratory and clinical perspective. ISRN surgery. 2014, 203943 (2014).
  25. de V. Alant, J. D., Kemp, S. W. P., Khu, K. J. O. L., Kumar, R., Webb, A. A., Midha, R. Traumatic neuroma in continuity injury model in rodents. Journal of Neurotrauma. 29 (8), 1691-1703 (2012).
  26. Geary, M. B., et al. Erythropoietin accelerates functional recovery after moderate sciatic nerve crush injury. Muscle & Nerve. 56 (1), 143-151 (2017).
  27. Răducan, A., et al. Morphological and functional aspects of sciatic nerve regeneration after crush injury. Romanian Journal of Morphology and Embryology. 54, 735-739 (2013).
  28. Elfar, J. C., Jacobson, J. A., Puzas, J. E., Rosier, R. N., Zuscik, M. J. Erythropoietin accelerates functional recovery after peripheral nerve injury. The Journal of Bone and Joint Surgery. American Volume. 90 (8), 1644-1653 (2008).
  29. Sundem, L., Chris Tseng, K. C., Li, H., Ketz, J., Noble, M., Elfar, J. Erythropoietin enhanced recovery after traumatic nerve injury: myelination and localized effects. The Journal of Hand Surgery. 41 (10), 999-1010 (2016).
  30. Govindappa, P. K., Talukder, M. A. H., Gurjar, A. A., Hegarty, J. P., Elfar, J. C. An effective erythropoietin dose regimen protects against severe nerve injury-induced pathophysiological changes with improved neural gene expression and enhances functional recovery. International Immunopharmacology. 82, 106330 (2020).
  31. Wandling, G. D., Lee, J. I., Talukder, M. A. H., Govindappa, P. K., Elfar, J. C. Novel real-time digital pressure sensor reveals wide variations in current nerve crush injury models. Military Medicine. 186, 473-478 (2021).
  32. Lee, J. I., Talukder, M. A. H., Karuman, K., Gurjar, A. A., Govindappa, P. K., Guddadarangaiah, J. M., Manto, K. M., Wandling, G. D., Hegarty, J. P., Waning, D. L., Elfar, J. C. Functional recovery and muscle atrophy in pre-clinical models of peripheral nerve transection and gap-grafting in mice. Effects of 4-aminopyridine. Neural Regeneration Research. , (2021).
  33. Lee, D. H., You, J., Jung, J. W., Park, J. W., Lee, J. I. Comparison between normal and reverse orientation of graft in functional and histomorphological outcomes after autologous nerve grafting: An experimental study in the mouse model. Microsurgery. 41 (7), 645-654 (2021).
  34. Lubiatowski, P., Unsal, F. M., Nair, D., Ozer, K., Siemionow, M. The epineural sleeve technique for nerve graft reconstruction enhances nerve recovery. Microsurgery. 28 (3), 160-167 (2008).
  35. Lee, J. I., Wandling, G. D., Talukder, M. A. H., Govindappa, P. K., Elfar, J. C. A novel standardized peripheral nerve transection method and a novel digital pressure sensor device construction for peripheral nerve crush injury. Bio-protocol. , (2022).
  36. Farinas, A. F., Stephanides, M., Kassis, S., Keller, P., Colazo, J. M., Thayer, W. P. Sciatic nerve injury model in rabbits: What to expect. Laboratory Animals. 54 (6), 559-567 (2020).
  37. Yao, Y., et al. Efect of longitudinally oriented collagen conduit combined with nerve growth factor on nerve regeneration after dog sciatic nerve injury. Journal of Biomedical Materials Research. Part B, Applied Biomaterials. 106 (6), 2131-2139 (2018).
  38. Schmitz, H. C., Beer, G. M. The toe-spreading reflex of the rabbit revisited–functional evaluation of complete peroneal nerve lesions. Laboratory Animals. 35 (4), 340-345 (2001).
  39. Bauder, A. R., Ferguson, T. A. Reproducible mouse sciatic nerve crush and subsequent assessment of regeneration by whole mount muscle analysis. Journal of Visualized Experiments. 60, 3606 (2012).
  40. Colleoni, M., Sacerdote, P. Murine models of human neuropathic pain. Biochimica et Biophysica Acta. 1802 (10), 924-933 (2010).
  41. Challa, S. R. Surgical animal models of neuropathic pain: Pros and Cons. The International Journal of Neuroscience. 125 (3), 170-174 (2015).
  42. M, F. G., M, M., S, H., Khan, W. S. Peripheral nerve injury: principles for repair and regeneration. The Open Orthopaedics Journal. 8, 199-203 (2014).
  43. Grinsell, D., Keating, C. P. Peripheral nerve reconstruction after injury: a review of clinical and experimental therapies. BioMed Research International. 2014, 698256 (2014).
  44. Carvalho, C. R., Reis, R. L., Oliveira, J. M. Fundamentals and current strategies for peripheral nerve repair and regeneration. Advances in Experimental Medicine and Biology. 1249, 173-201 (2020).
  45. Govindappa, P. K., Jagadeeshaprasad, M. G., Tortora, P., Talukder, M. A. H., Elfar, J. C. Effects of 4-aminopyridine on combined nerve and muscle injury and bone loss. The Journal of Hand Surgery. , (2022).
This article has been published
Video Coming Soon
Keep me updated:

.

Cite This Article
Lee, J. I., Govindappa, P. K., Wandling, G. D., Elfar, J. C. Traumatic Peripheral Nerve Injury in Mice. J. Vis. Exp. (181), e63551, doi:10.3791/63551 (2022).

View Video