Summary

Establishment of a Robust and Reproducible Model of Radiation-Induced Skin and Muscle Fibrosis

Published: August 31, 2022
doi:

Summary

Here we present a protocol to induce radiation-induced skin fibrosis in the hind limb of mice and perform post-irradiation measurements of chronic impairment via limb excursion and gait index analyses to evaluate the functional outcome. The model elucidates radiation-related skin fibrosis mechanisms and is useful in subclinical therapeutic studies.

Abstract

Radiation-induced skin fibrosis (RISF) can result from a plethora of scenarios including cancer therapy, accidental exposure, or acts of terrorism. Radioactive beams can penetrate through the skin and affect the structures in their path including skin, muscles, and internal organs. Skin is the first structure to get exposed to radiation and is susceptible to develop chronic fibrosis, which is challenging to treat. Currently, limited treatment options show moderate efficacy in mitigating radiation-related skin fibrosis. A key factor hindering the development of effective countermeasures is the absence of a convenient and robust model that could allow for translation of the experimental findings to humans. Here, a robust and reproducible murine hind limb skin fibrosis model has been established for prophylactic and therapeutic evaluation of possible agents for functional and molecular recovery.

The right hind limb was irradiated using a single dose of 40 (Gray) Gy to induce skin fibrosis. Subjects developed edema and dermatitis in the early stages proceeded by visible skin constriction. Irradiated limbs showed a significantly reduced limb range of motion in the following weeks. In late stages, acute side effects subsided, yet chronic fibrosis persisted. A gait index was performed as an additional functional assay, which demonstrated the development of functional impairment. These non-invasive methods demonstrated reliable measurements for tracing fibrosis progression, which is supported by histological analyses. The radiation dose, application, and post-irradiation analyses employed in this model offer a vigorous and reproducible method for studying radiation-induced skin fibrosis and testing the efficacy of therapeutical agents.

Introduction

The skin is the largest organ of the body, covering and protecting the body from hazards. It has three distinct layers: epidermis, dermis, and hypodermis. Each layer has its unique functions: the epidermis prevents dehydration and microbial invasion; the dermis has a rich network of cells, and an extracellular matrix that provides tensile strength and elasticity1; the dermal layer contains the sensory receptors, hair follicles, glands, and vessels for lymphatic and capillary networks. The hypodermis or subcutaneous tissue, with its abundance of adipose tissue, contours the body and distributes mechanical stress2,3,4.

Radiation, generated as a result of accidents, war, terrorism, or therapeutical applications, penetrate through the body in a linear progressive nature, leading the skin to be the first organ to come in contact. The threat of such incidents has intensified due to the increased use of radioactive materials in industries, medical facilities, and military installations5. Clinically, radiation damage to the skin is characterized by cutaneous radiation syndrome (CRS), one of four sub-syndromes of acute radiation syndrome (ARS). The response of the skin to ionizing radiation has important implications for treatment and protection from further damage6. Concomitant injuries such as burns and trauma further complicate the clinical outcome when combined with radiation injuries7. The extent of skin exposure to radiation correlates to a point-of-no-return threshold, from which the impairment of other organs results in single or multiple organ failure, and ultimately leads to patient death8,9. Cutaneous radiation injury is comprised of an acute and a chronic phase. Acute radiation injury clinically manifests as erythema, skin edema, dermatitis, blistering, epidermal denudation, dry or wet desquamation, ulceration, and changes in the hair and nails. The chronic phase is manifested as dermal atrophy, fibrosis, chronic ulceration, and telangiectasias10,11. In general, acute effects are predominately manifested in the epidermis, while chronic effects are most prominent in the dermis. Acute reaction to radiation exposure leads to a marked decrease in mitotic activity within 12 h of exposure, followed by hyperemia, cell enlargement, vacuolization, nuclear pyknosis, and fragmentation4,12.

Radiation doses exceeding 40 Gy result in moist desquamation and loss of epidermis, leading to an increased susceptibility to infections13. In addition, skin exposure to radiation induces cytokine production, triggering an inflammatory immune response in the dermal layer. Prominent inflammatory mediators include interleukins (IL-1, IL-3, IL-5, IL-6, and IL-8) and tumor necrosis factor-α (TNFα)14. Failure in the resolution of inflammation can eventually result in fibrosis development at the site of radiation injury15. Additional physical wounds or thermal injuries further aggravate this fibrotic response, extending through the muscle layer16. Transforming growth factor-β (TGFβ) is the key cytokine in fibrosis development17. Currently, very few treatment options show promising results, and the majority might have challenges with patient compliance. Further research exploring the cellular and molecular responses of the skin to different radiation doses will improve the understanding of the radiation-induced skin pathophysiology and enhance the development of new therapies.

To facilitate the clinical translation of research outcomes in preclinical models in alleviating radiation-induced injury to the skin and soft tissues, designing highly relevant experimental models of therapeutic interventions following irradiation is crucial. Both in vitro and in vivo models of radiation-induced injury have been described, including cell culture models of irradiated endothelial cells18,19, fibroblasts20, or keratinocytes19 and in vivo rodent, swine, and non-human primate animal models. Rodent models are widely used in radiation research due to their similarities in response to radiation injury with humans and their flexibility of genetic manipulation21. Radiation dose requirements are higher in rodents than in humans when seeking similar outcomes: desquamation, fibrosis, and necrosis16,22. Description of scoring criteria to measure the response to radiation has further enhanced the adoption of rodent models of radiation skin injury21,23.

Current research in the preclinical setting focuses on understanding the mechanisms of radiation-induced skin injury and developing therapeutical options. Thus, establishing a robust and reproducible preclinical model to create the radiation insult with high clinical translatability is essential. This work describes a murine model of skin fibrosis with optimized radiation dose and delivery technique. Our model, which combines functional, histological, and molecular measurements, can be used to effectively study the mechanism of fibrosis development and investigate new therapeutical options.

Protocol

Ethical animal use was approved by the Institutional Animal Care and Use Committee (IACUC), which acts in compliance with the Animal Welfare Act. Animals were housed in an Association for Assessment and Accreditation of Laboratory Animal Care International (AAALAC) approved facility and treated according to the National Institutes of Health Guide for the Care and Use of Laboratory Animals. 1. Anesthesia Place mice in the box of a small animal anesthesia system (<st…

Representative Results

Establishment and optimization of the current irradiation protocol resulted in a consistent and reproducible induction of fibrosis in mice. Right limbs of the mice were positioned and secured within the radiation field on the day of irradiation, and 40 Gy of radiation was administered. The development of functional impairment in skin was monitored by capturing images every week, post-irradiation. Photos showed that the optimized protocol created fibrosis by day 40 with 95% confidence. An examp…

Discussion

Skin injury is a likely outcome of accidental or medical treatment-related exposure to radiation. Nuclear reactors possess an accidental breach risk due to human error or natural disasters like Chernobyl and Fukushima26,27. Therapeutical dosing for cancer treatment is the most common exposure, which uses fractionated repeated dose regimens that risk causing radiation-related fibrosis in the treated areas. This common chronic adverse reaction can be prevalent in u…

Divulgazioni

The authors have nothing to disclose.

Acknowledgements

This work is funded by research grants from the Department of Defense W81XWH-19-PRMRP-DA, NIAID/NIH Grant 5R21AI153971-02, and PSF/MTF Grant 603902.

Materials

10% Formalin Fischer Scientific 23-427098
Bolus Orfit 8333.SO1/R
Clipper Kent Scientific Corp. CL8787-KIT
CO2 Various
CO2 Chamber E-Z Systems Inc. E-22000
Depilatory Cream Church & Dwight Co., Inc. Nair
Digital Camera Wolfang GA100
Eppendrof Tubes Eppendorf 22364111
Eye Lubricant Dechra Puralube Ophthalmic Ointment
Gauze Covidien 682252
Image Processing Program NIH Image J
Isoflurane Dechra USP Inhalation Anesthetic
Linear Accelaerator Varian Medical Systems, Inc. 23EX
PBS Cytiva SH30256.LS
Pentobarbital Akorn Pharmaceuticals Nembutal
Protractor Westcott 550-1120
Small Animal Anesthesia System E-Z Systems Inc. EZ-SA800 Single animal system
Spreadsheet Software Microsoft Excel
Surgical Scissors Medline MDS0834111
Surgical Tape 3M 1538-1
Tape 3M H-1113

Riferimenti

  1. Breitkreutz, D., Mirancea, N., Nischt, R. Basement membranes in skin: unique matrix structures with diverse functions. Histochemistry and Cell Biology. 132 (1), 1-10 (2009).
  2. Kim, J. -. S., et al. Comparison of skin injury induced by β-and γ-irradiation in the minipig model. Journal of Radiation Protection and Research. 42 (4), 189-196 (2017).
  3. Kolarsick, P. A., Kolarsick, M. A., Goodwin, C. Anatomy and physiology of the skin. Journal of the Dermatology Nurses’ Association. 3 (4), 203-213 (2011).
  4. von Essen, C. F. Radiation tolerance of the skin. Acta Radiologica: Therapy, Physics, Biology. 8 (4), 311-330 (1969).
  5. Dainiak, N., et al. Literature review and global consensus on management of acute radiation syndrome affecting nonhematopoietic organ systems. Disaster Medicine and Public Health Preparedness. 5 (3), 183-201 (2011).
  6. Hopewell, J. The skin: its structure and response to ionizing radiation. International Journal of Radiation Biology. 57 (4), 751-773 (1990).
  7. Flynn, D. F., Goans, R. E. Nuclear terrorism: triage and medical management of radiation and combined-injury casualties. Surgical Clinics. 86 (3), 601-636 (2006).
  8. Peter, R. Cutaneous radiation syndrome in multi-organ failure. The British Journal of Radiology. 78 (1), 180-184 (2005).
  9. Meineke, V. The role of damage to the cutaneous system in radiation-induced multi-organ failure. The British Journal of Radiology. 78 (1), 95-99 (2005).
  10. Berger, M., Christensen, D., Lowry, P., Jones, O., Wiley, A. Medical management of radiation injuries: current approaches. Occupational Medicine. 56 (3), 162-172 (2006).
  11. Ralf, U. P., Petra, G. Management of cutaneous radiation injuries: diagnostic and therapeutic principles of the cutaneous radiation syndrome. Military Medicine. 167, 110-112 (2002).
  12. Bray, F. N., Simmons, B. J., Wolfson, A. H., Nouri, K. Acute and chronic cutaneous reactions to ionizing radiation therapy. Dermatology and Therapy. 6 (2), 185-206 (2016).
  13. Mendelsohn, F. A., Divino, C. M., Reis, E. D., Kerstein, M. D. Wound care after radiation therapy. Advances in Skin & Wound. 15 (5), 216-224 (2002).
  14. Peter, R. U. . Radiation Treatment and Radiation Reactions in Dermatology. , 185-188 (2015).
  15. Ejaz, A., Greenberger, J. S., Rubin, P. J. Understanding the mechanism of radiation induced fibrosis and therapy options. Pharmacology & Therapeutics. 204, 107399 (2019).
  16. Williams, J. P., et al. Animal models for medical countermeasures to radiation exposure. Radiation research. 173 (4), 557-578 (2010).
  17. Ejaz, A., Epperly, M. W., Hou, W., Greenberger, J. S., Rubin, J. P. Adipose-derived stem cell therapy ameliorates ionizing irradiation fibrosis via hepatocyte growth factor-mediated transforming growth factor-beta downregulation and recruitment of bone marrow cells. Stem Cells. 37 (6), 791-802 (2019).
  18. Haubner, F., et al. Effects of external radiation in a co-culture model of endothelial cells and adipose-derived stem cells. Radiation Oncology. 8 (1), 66 (2013).
  19. Ebrahimian, T. G., et al. Cell therapy based on adipose tissue-derived stromal cells promotes physiological and pathological wound healing. Arteriosclerosis, Thrombosis, and Vascular Biology. 29 (4), 503-510 (2009).
  20. Haubner, F., et al. A co-culture model of fibroblasts and adipose tissue-derived stem cells reveals new insights into impaired wound healing after radiotherapy. International Journal of Molecular Sciences. 16 (11), 25947-25958 (2015).
  21. Urano, M., Kenton, L. A., Kahn, J. The effect of hyperthermia on the early and late appearing mouse foot reactions and on the radiation carcinogenesis: effect on the early and late appearing reactions. International Journal of Radiation Oncology Biology Physics. 15 (1), 159-166 (1988).
  22. Law, M., Thomlinson, R. The pathogenesis of necrosis after radiotherapy. The British Journal of Radiology. 47 (562), 740 (1974).
  23. Abe, Y., Urano, M. Fraction size-dependent acute skin reaction of mice after multiple twice-a-day doses. International Journal of Radiation Oncology Biology Physics. 18 (2), 359-364 (1990).
  24. Inserra, M. M., Bloch, D. A., Terris, D. J. Functional indices for sciatic, peroneal, and posterior tibial nerve lesions in the mouse. Microsurgery. 18 (2), 119-124 (1998).
  25. Suckow, C. P. S. a. M. A., Weichbrod, R. H., Thompson, G. A., Norton, J. N. . Management of Animal Care and Use Programs in Research, Education, and Testing. , (2018).
  26. Yamashita, S., Suzuki, S., Suzuki, S., Shimura, H., Saenko, V. Lessons from Fukushima: latest findings of thyroid cancer after the Fukushima nuclear power plant accident. Thyroid. 28 (1), 11-22 (2018).
  27. Cardis, E., et al. Cancer consequences of the Chernobyl accident: 20 years on. Journal of Radiological Protection. 26 (2), 127-140 (2006).
  28. Williams, N. R., et al. Radiation-induced fibrosis in breast cancer: A protocol for an observational cross-sectional pilot study for personalised risk estimation and objective assessment. International Journal of Surgery Protocols. 14, 9-13 (2019).
  29. Meineke, V., Fliedner, T. Radiation-induced multi-organ involvement and failure: challenges for radiation accident medical management and future research. The British Journal of Radiology. 78 (1), 196-200 (2005).
  30. Stone, H. B. Leg contracture in mice: an assay of normal tissue response. International Journal of Radiation Oncology Biology Physics. 10 (7), 1053-1061 (1984).
This article has been published
Video Coming Soon
Keep me updated:

.

Citazione di questo articolo
Surucu, Y., Bengur, F. B., Yang, K. S., Schilling, B. K., Baker, J. S., Shabbir, S., Fisher, R., Epperly, M. W., Greenberger, J. S., Rubin, J. P., Ejaz, A. Establishment of a Robust and Reproducible Model of Radiation-Induced Skin and Muscle Fibrosis. J. Vis. Exp. (186), e64251, doi:10.3791/64251 (2022).

View Video