What a pain in the back: etiology, diagnosis and future treatment directions for discogenic low back pain

Giselle Kaneda , Lea Zila , Jacob T. Wechsler , Karim Shafi , Karandeep Cheema , Hyun Bae , Sang D. Kim , Alexander Tuchman , Debiao Li , Dmitriy Sheyn

Bone Research ›› 2025, Vol. 13 ›› Issue (1) : 89

PDF
Bone Research ›› 2025, Vol. 13 ›› Issue (1) : 89 DOI: 10.1038/s41413-025-00472-7
Review Article
review-article

What a pain in the back: etiology, diagnosis and future treatment directions for discogenic low back pain

Author information +
History +
PDF

Abstract

Chronic lower back pain (LBP) is the leading cause of disability worldwide. Due to its close relationship with intervertebral disc (IVD) degeneration (IVDD), research has historically focused more on understanding the mechanism behind IVDD while clinical efforts prioritize pain management. More recently, there has been a shift toward understanding LBP as a distinct pathological entity. This review synthesizes current knowledge on discogenic LBP, combining known pathophysiology, molecular mechanisms, risk factors, diagnostic challenges, and available experimental models. IVDD is a complex, multifactorial process involving biochemical, mechanical, and inflammatory changes within the disc, leading to structural breakdown and potential discogenic pain. Key mechanisms include extracellular matrix degradation, upregulation of inflammatory mediators, immune cell infiltration, and aberrant nerve and vascular ingrowth. However, not all cases of IVDD result in LBP, highlighting the need for further investigation into the cellular, molecular, and biomechanical factors contributing to symptom development. Current diagnostic tools and experimental models for studying discogenic LBP remain limited, impeding the development of targeted treatments. Existing therapies primarily focus on symptom management rather than addressing underlying disease mechanisms.

Cite this article

Download citation ▾
Giselle Kaneda, Lea Zila, Jacob T. Wechsler, Karim Shafi, Karandeep Cheema, Hyun Bae, Sang D. Kim, Alexander Tuchman, Debiao Li, Dmitriy Sheyn. What a pain in the back: etiology, diagnosis and future treatment directions for discogenic low back pain. Bone Research, 2025, 13(1): 89 DOI:10.1038/s41413-025-00472-7

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Collaborators GBDLBP. Global, regional, and national burden of low back pain, 1990-2020, its attributable risk factors, and projections to 2050: a systematic analysis of the global burden of didisease study 2021. Lancet Rheumatol., 2023, 5: e316-e329.

[2]

Simon J, McAuliffe M, Shamim F, Vuong N, Tahaei A. Discogenic low back pain. Phys. Med Rehabil. Clin. N. Am., 2014, 25: 305-317.

[3]

Jiang W. et al.. Intervertebral disc human nucleus pulposus cells associated with back pain trigger neurite outgrowth in vitro and pain behaviors in rats. Sci. Transl. Med., 2023, 15. eadg7020

[4]

Melrose, J. & Roughley, P. in The Intervertebral Disc: Molecular and Structural Studies of the Disc in Health and Disease (eds Irving M. Shapiro & Makarand V. Risbud) 53–77 (Springer Vienna, 2014).

[5]

Dou Y, Sun X, Ma X, Zhao X, Yang Q. Intervertebral disk degeneration: the microenvironment and tissue engineering strategies. Front. Bioeng. Biotechnol., 2021, 9. 592118

[6]

Rider SM, Mizuno S, Kang JD. olecular mechanisms of intervertebral disc degeneration. Spine Surg. Relat. Res., 2019, 3: 1-11.

[7]

Waxenbaum, J. A., Reddy, V. & Futterman, B. Anatomy, Back, Intervertebral Discs. in StatPearls [Internet]. (StatPearls Publishing, Treasure Island (FL), 2023). Available from: https://www.ncbi.nlm.nih.gov/books/NBK470583/.

[8]

Mohd Isa, I. L., Teoh, S. L., Mohd Nor, N. H. & Mokhtar, S. A. Discogenic low back pain: anatomy, pathophysiology and treatments of intervertebral disc degeneration. Int. J. Mol. Sci.24, 208 (2022).

[9]

Fields AJ, Ballatori A, Liebenberg EC, Lotz JC. Contribution of the endplates to disc degeneration. Curr. Mol. Biol. Rep., 2018, 4: 151-160.

[10]

Lotz JC, Fields AJ, Liebenberg EC. The role of the vertebral end plate in low back pain. Glob. Spine J., 2013, 3: 153-164.

[11]

Boos N. et al.. Classification of age-related changes in lumbar intervertebral discs: 2002 Volvo Award in basic science. Spine (Philos. Pa 1976), 2002, 27: 2631-2644.

[12]

An HS, Masuda K, Inoue N. Intervertebral disc degeneration: biological and biomechanical factors. J. Orthop. Sci., 2006, 11: 541-552.

[13]

Antoniou J. et al.. The human lumbar intervertebral disc: evidence for changes in the biosynthesis and denaturation of the extracellular matrix with growth, maturation, ageing, and degeneration. J. Clin. Invest., 1996, 98: 996-1003.

[14]

Alkhatib B. et al.. Acute mechanical injury of the human intervertebral disc: link to degeneration and pain. Eur. Cell Mater., 2014, 28: 98-110.

[15]

Zhao CQ, Wang LM, Jiang LS, Dai LY. The cell biology of intervertebral disc aging and degeneration. Ageing Res Rev., 2007, 6: 247-261.

[16]

Xiao H, Wang K, Peng L, Yin Z. Laquinimod attenuates oxidative stress-induced mitochondrial injury and alleviates intervertebral disc degeneration by inhibiting the NF-kappaB signaling pathway. Int. Immunopharmacol., 2024, 131. 111804

[17]

Vergroesen PP. et al.. Mechanics and biology in intervertebral disc degeneration: a vicious circle. Osteoarthr. Cartil., 2015, 23: 1057-1070.

[18]

Yang H. et al.. Secreted factors from intervertebral disc cells and infiltrating macrophages promote degenerated intervertebral disc catabolism. Spine (Philos. Pa 1976), 2019, 44: E520-E529.

[19]

Sivan SS. et al.. Biochemical composition and turnover of the extracellular matrix of the normal and degenerate intervertebral disc. Eur. Spine J., 2014, 23: S344-S353.

[20]

Vo NV. et al.. Expression and regulation of metalloproteinases and their inhibitors in intervertebral disc aging and degeneration. Spine J., 2013, 13: 331-341.

[21]

Zhang Y. et al.. Melatonin modulates IL-1beta-induced extracellular matrix remodeling in human nucleus pulposus cells and attenuates rat intervertebral disc degeneration and inflammation. Aging (Albany NY), 2019, 11: 10499-10512.

[22]

Panebianco CJ, Dave A, Charytonowicz D, Sebra R, Iatridis JC. Single-cell RNA-sequencing atlas of bovine caudal intervertebral discs: discovery of heterogeneous cell populations with distinct roles in homeostasis. FASEB J., 2021, 35. e21919

[23]

Rohanifar M. et al.. Single cell RNA-sequence analyses reveal uniquely expressed genes and heterogeneous immune cell involvement in the rat model of intervertebral disc degeneration. Appl. Sci, 2022, 12: 8244.

[24]

Wang D. et al.. Single-cell transcriptomics reveals heterogeneity and intercellular crosstalk in human intervertebral disc degeneration. iScience, 2023, 26. 106692

[25]

Später T. et al.. Retention of human iPSC-derived or primary cells following xenotransplantation into rat immune-privileged sites. Bioengineering, 2023, 10: 1049.

[26]

Molinos M. et al.. Inflammation in intervertebral disc degeneration and regeneration. J. R. Soc. Interface, 2015, 12: 20141191.

[27]

Johnson ZI, Schoepflin ZR, Choi H, Shapiro IM, Risbud MV. Disc in flames: roles of TNF-alpha and IL-1beta in intervertebral disc degeneration. Eur. Cell Mater., 2015, 30: 104-116.

[28]

Liang CZ. et al.. The relationship between low pH in intervertebral discs and low back pain: a systematic review. Arch. Med. Sci., 2012, 8: 952-956.

[29]

Bez M. et al.. Molecular pain markers correlate with pH-sensitive MRI signal in a pig model of disc degeneration. Sci. Rep., 2018, 8. 17363

[30]

Sheyn D. et al.. Human iPSCs can be differentiated into notochordal cells that reduce intervertebral disc degeneration in a porcine model. Theranostics, 2019, 9: 7506-7524.

[31]

Zhou Z. et al.. Quantitative chemical exchange saturation transfer MRI of intervertebral disc in a porcine model. Magn. Reson. Med., 2016, 76: 1677-1683.

[32]

Hartvigsen J. et al.. What low back pain is and why we need to pay attention. Lancet, 2018, 391: 2356-2367.

[33]

Maher C, Underwood M, Buchbinder R. Non-specific low back pain. Lancet, 2017, 389: 736-747.

[34]

Henschke N. et al.. Prevalence of and screening for serious spinal pathology in patients presenting to primary care settings with acute low back pain. Arthritis Rheum., 2009, 60: 3072-3080.

[35]

Trainor TJ, Trainor MA. Etiology of low back pain in athletes. Curr. Sports Med. Rep., 2004, 3: 41-46.

[36]

Middleton K, Fish DE. Lumbar spondylosis: clinical presentation and treatment approaches. Curr. Rev. Musculoskelet. Med, 2009, 2: 94-104.

[37]

Walter KL, O’Toole JE. Lumbar spinal stenosis. JAMA, 2022, 328: 310.

[38]

Awadalla AM. et al.. Management of lumbar disc herniation: a systematic review. Cureus, 2023, 15: e47908

[39]

Schwarzer AC. et al.. The relative contributions of the disc and zygapophyseal joint in chronic low back pain. Spine (Philos. Pa 1976), 1994, 19: 801-806.

[40]

de Schepper EI. et al.. The association between lumbar disc degeneration and low back pain: the influence of age, gender, and individual radiographic features. Spine (Philos. Pa 1976), 2010, 35: 531-536.

[41]

Brinjikji W. et al.. MRI findings of disc degeneration are more prevalent in adults with low back pain than in asymptomatic controls: a systematic review and meta-analysis. AJNR Am. J. Neuroradiol., 2015, 36: 2394-2399.

[42]

Groh AMR, Fournier DE, Battie MC, Seguin CA. Innervation of the human intervertebral disc: a scoping review. Pain. Med., 2021, 22: 1281-1304.

[43]

Ratsep T, Minajeva A, Asser T. Relationship between neovascularization and degenerative changes in herniated lumbar intervertebral discs. Eur. Spine J., 2013, 22: 2474-2480.

[44]

Xu J, Shao T, Lou J, Zhang J, Xia C. Aging, cell senescence, the pathogenesis and targeted therapies of intervertebral disc degeneration. Front. Pharm., 2023, 14. 1172920

[45]

Dowdell J. et al.. Intervertebral disk degeneration and repair. Neurosurgery, 2017, 80: S46-S54.

[46]

Jha, R. et al. Updates on pathophysiology of discogenic back pain. J. Clin. Med.12, 6907 (2023).

[47]

De Simone, M. et al. Discogenic low back pain: anatomic and pathophysiologic characterization, clinical evaluation, biomarkers, AI, and treatment options. J. Clin. Med.13, 5915 (2024).

[48]

Chiu AP. et al.. Human molecular mechanisms of discogenic low back pain: a scoping review. J. Pain., 2025, 27. 104693

[49]

Cooke PM, Lutz GE. Internal disc disruption and axial back pain in the athlete. Phys. Med Rehabil. Clin. N. Am., 2000, 11: 837-865.

[50]

Deyo RA, Weinstein JN. Low back pain. N. Engl. J. Med., 2001, 344: 363-370.

[51]

Koes BW, van Tulder MW, Ostelo R, Kim Burton A, Waddell G. Clinical guidelines for the management of low back pain in primary care: an international comparison. Spine (Philos. Pa 1976), 2001, 26: 2504-2513.

[52]

Wang, J. C., Lamartina, C. & AOSpine International (Firm). AOSpine masters series. Volume 8, Back pain. (Thieme, 2017).

[53]

Fujii, K. et al. Discogenic back pain: literature review of definition, diagnosis, and treatment. JBMR 3, e10180 (2019).

[54]

Bridwell, K. H., Bridwell, K. H. & Gupta, M. C. Bridwell and DeWald’s textbook of spinal surgery. Fourth edition edn, (Wolters Kluwer Health, 2019).

[55]

Boden SD, Davis DO, Dina TS, Patronas NJ, Wiesel SW. Abnormal magnetic-resonance scans of the lumbar spine in asymptomatic subjects. A prospective investigation. J. Bone Jt. Surg. Am., 1990, 72: 403-408.

[56]

Jensen MC. et al.. Magnetic resonance imaging of the lumbar spine in people without back pain. N. Engl. J. Med, 1994, 331: 69-73.

[57]

Stadnik TW. et al.. Annular tears and disk herniation: prevalence and contrast enhancement on MR images in the absence of low back pain or sciatica. Radiology, 1998, 206: 49-55.

[58]

Weishaupt D, Zanetti M, Hodler J, Boos N. MR imaging of the lumbar spine: prevalence of intervertebral disk extrusion and sequestration, nerve root compression, end plate abnormalities, and osteoarthritis of the facet joints in asymptomatic volunteers. Radiology, 1998, 209: 661-666.

[59]

Jarvik JJ, Hollingworth W, Heagerty P, Haynor DR, Deyo RA. The Longitudinal Assessment of Imaging and Disability of the Back (LAIDBack) Study: baseline data. Spine (Philos. Pa 1976), 2001, 26: 1158-1166.

[60]

Pfirrmann CW, Metzdorf A, Zanetti M, Hodler J, Boos N. Magnetic resonance classification of lumbar intervertebral disc degeneration. Spine (Philos. Pa 1976), 2001, 26: 1873-1878.

[61]

Brayda-Bruno M. et al.. Advances in the diagnosis of degenerated lumbar discs and their possible clinical application. Eur. Spine J., 2014, 23: S315-S323.

[62]

Schellhas KP, Pollei SR, Gundry CR, Heithoff KB. Lumbar disc high-intensity zone. Correlation of magnetic resonance imaging and discography. Spine (Philos. Pa 1976), 1996, 21: 79-86.

[63]

Marinelli NL, Haughton VM, Munoz A, Anderson PA. T2 relaxation times of intervertebral disc tissue correlated with water content and proteoglycan content. Spine (Philos. Pa 1976), 2009, 34: 520-524.

[64]

Marinelli NL, Haughton VM, Anderson PA. T2 relaxation times correlated with stage of lumbar intervertebral disk degeneration and patient age. AJNR Am. J. Neuroradiol., 2010, 31: 1278-1282.

[65]

Blumenkrantz G. et al.. In vivo 3.0-tesla magnetic resonance T1rho and T2 relaxation mapping in subjects with intervertebral disc degeneration and clinical symptoms. Magn. Reson. Med., 2010, 63: 1193-1200.

[66]

Johannessen W. et al.. Assessment of human disc degeneration and proteoglycan content using T1rho-weighted magnetic resonance imaging. Spine (Philos. Pa 1976), 2006, 31: 1253-1257.

[67]

Vaga S. et al.. Quantitative assessment of intervertebral disc glycosaminoglycan distribution by gadolinium-enhanced MRI in orthopedic patients. Magn. Reson. Med., 2008, 59: 85-95.

[68]

Shapiro EM, Borthakur A, Gougoutas A, Reddy R. 23Na MRI accurately measures fixed charge density in articular cartilage. Magn. Reson. Med., 2002, 47: 284-291.

[69]

Lindblom K. Diagnostic puncture of intervertebral disks in sciatica. Acta Orthop. Scand., 1948, 17: 231-239.

[70]

Walker JIII., El Abd O, Isaac Z, Muzin S. Discography in practice: a clinical and historical review. Curr. Rev. Musculoskelet. Med., 2008, 1: 69-83.

[71]

Carragee EJ. et al.. The rates of false-positive lumbar discography in select patients without low back symptoms. Spine (Philos. Pa 1976), 2000, 25: 1373-1380.

[72]

Willems P. Decision making in surgical treatment of chronic low back pain: the performance of prognostic tests to select patients for lumbar spinal fusion. Acta Orthop. Suppl., 2013, 84: 1-35.

[73]

Eck JC. et al.. Guideline update for the performance of fusion procedures for degenerative disease of the lumbar spine. Part 6: discography for patient selection. J. Neurosurg. Spine, 2014, 21: 37-41.

[74]

Bizzoca D. et al.. Gender-related issues in the management of low-back pain: a current concepts review. Clin. Pr., 2023, 13: 1360-1368.

[75]

Wang YX, Wang JQ, Kaplar Z. Increased low back pain prevalence in females than in males after menopause age: evidences based on synthetic literature review. Quant. Imaging Med. Surg., 2016, 6: 199-206.

[76]

Bonnheim NB. et al.. ISSLS Prize in Bioengineering Science 2023: age- and sex-related differences in lumbar intervertebral disc degeneration between patients with chronic low back pain and asymptomatic controls. Eur. Spine J., 2023, 32: 1517-1524.

[77]

Lou C. et al.. Association between menopause and lumbar disc degeneration: an MRI study of 1566 women and 1382 men. Menopause, 2017, 24: 1136-1144.

[78]

Mosley GE. et al.. Sex differences in rat intervertebral disc structure and function following annular puncture injury. Spine (Philos. Pa 1976), 2019, 44: 1257-1269.

[79]

Mosley GE. et al.. Males and females exhibit distinct relationships between intervertebral disc degeneration and pain in a rat model. Sci. Rep., 2020, 10. 15120

[80]

Battie MC, Videman T, Levalahti E, Gill K, Kaprio J. Genetic and environmental effects on disc degeneration by phenotype and spinal level: a multivariate twin study. Spine (Philos. Pa 1976), 2008, 33: 2801-2808.

[81]

Livshits G. et al.. Lumbar disc degeneration and genetic factors are the main risk factors for low back pain in women: the UK Twin Spine Study. Ann. Rheum. Dis., 2011, 70: 1740-1745.

[82]

Sambrook PN, MacGregor AJ, Spector TD. Genetic influences on cervical and lumbar disc degeneration: a magnetic resonance imaging study in twins. Arthritis Rheum., 1999, 42: 366-372.

[83]

Zorkoltseva, I. V. et al. Multi-trait exome-wide association study of back pain-related phenotypes. Genes14, 1962 (2023).

[84]

Suri P. et al.. Genome-wide meta-analysis of 158 000 individuals of European ancestry identifies three loci associated with chronic back pain. PLoS Genet., 2018, 14: e1007601.

[85]

Zhang H. et al.. Association of single nucleotide polymorphism rs2228570 with lumbar disc degeneration: a case-control study and meta-analysis. J. Pain. Res., 2021, 14: 2001-2012.

[86]

Cauci S. et al.. Low back pain and FokI (rs2228570) polymorphism of vitamin D receptor in athletes. BMC Sports Sci. Med Rehabil., 2017, 9: 4.

[87]

Baumbauer KM. et al.. Contribution of COMT and BDNF genotype and expression to the risk of transition from acute to chronic low back pain. Clin. J. Pain., 2020, 36: 430-439.

[88]

Martirosyan NL. et al.. Genetic alterations in intervertebral disc disease. Front. Surg., 2016, 3: 59.

[89]

Solovieva S. et al.. Possible association of interleukin 1 gene locus polymorphisms with low back pain. Pain, 2004, 109: 8-19.

[90]

Biczo A. et al.. Genetic variants of interleukin 1B and 6 are associated with clinical outcome of surgically treated lumbar degenerative disc disease. BMC Musculoskelet. Disord., 2022, 23. 774

[91]

Trompeter K, Fett D, Platen P. Prevalence of back pain in sports: a systematic review of the literature. Sports Med, 2017, 47: 1183-1207.

[92]

Sweeney EA, Daoud AK, Potter MN, Ritchie L, Howell DR. Association between flexibility and low back pain in female adolescent gymnasts. Clin. J. Sport Med., 2019, 29: 379-383.

[93]

Oliva FA, Vitaterna MN, Maffulli MCM. Low back pain in weightlifters: personalised exercise protocols for elite athletes. Muscles, Ligaments Tendons J., 2023, 13: 187-200.

[94]

Nugent, F. J. et al. The relationship between rowing-related low back pain and rowing biomechanics: a systematic review. Br J. Sports Med. (2021). https://doi.org/10.1136/bjsports-2020-102533

[95]

Murillo C. et al.. High-density electromyography provides new insights into the flexion relaxation phenomenon in individuals with low back pain. Sci. Rep., 2019, 9. 15938

[96]

Baradaran Mahdavi S, Riahi R, Vahdatpour B, Kelishadi R. Association between sedentary behavior and low back pain; a systematic review and meta-analysis. Health Promot Perspect., 2021, 11: 393-410.

[97]

da Cruz Fernandes IM, Pinto RZ, Ferreira P, Lira FS. Low back pain, obesity, and inflammatory markers: exercise as potential treatment. J. Exerc Rehabil., 2018, 14: 168-174.

[98]

Wong AYL, Karppinen J, Samartzis D. Low back pain in older adults: risk factors, management options and future directions. Scoliosis Spinal Disord., 2017, 12. 14

[99]

Wang Y. et al.. The role of IL-1beta and TNF-alpha in intervertebral disc degeneration. Biomed. Pharmacother., 2020, 131. 110660

[100]

Hise AG. et al.. An essential role for the NLRP3 inflammasome in host defense against the human fungal pathogen Candida albicans. Cell Host Microbe, 2009, 5: 487-497.

[101]

Mariathasan S. et al.. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature, 2006, 440: 228-232.

[102]

Ma M, Jiang W, Zhou R. DAMPs and DAMP-sensing receptors in inflammation and diseases. Immunity, 2024, 57: 752-771.

[103]

Ohnishi, T., Iwasaki, N. & Sudo, H. Causes of and molecular targets for the treatment of intervertebral disc degeneration: a review. Cells11, 394 (2022).

[104]

Dela Cruz CS, Kang MJ. Mitochondrial dysfunction and damage associated molecular patterns (DAMPs) in chronic inflammatory diseases. Mitochondrion, 2018, 41: 37-44.

[105]

Nakahira K, Hisata S, Choi AM. The roles of mitochondrial damage-associated molecular patterns in diseases. Antioxid. Redox Signal, 2015, 23: 1329-1350.

[106]

Murphy, K., Lufkin, T. & Kraus, P. Development and degeneration of the intervertebral disc-Insights from across species. Vet. Sci.10, 540 (2023).

[107]

Quero L. et al.. Hyaluronic acid fragments enhance the inflammatory and catabolic response in human intervertebral disc cells through modulation of toll-like receptor 2 signalling pathways. Arthritis Res. Ther., 2013, 15. R94

[108]

Schmidli MR. et al.. Fibronectin fragments and inflammation during canine intervertebral disc disease. Front. Vet. Sci., 2020, 7. 547644

[109]

Roh JS, Sohn DH. Damage-associated molecular patterns in inflammatory diseases. Immune Netw., 2018, 18. e27

[110]

Klatt AR. et al.. A critical role for collagen II in cartilage matrix degradation: collagen II induces pro-inflammatory cytokines and MMPs in primary human chondrocytes. J. Orthop. Res., 2009, 27: 65-70.

[111]

Ruettger A, Schueler S, Mollenhauer JA, Wiederanders B. Cathepsins B, K, and L are regulated by a defined collagen type II peptide via activation of classical protein kinase C and p38 MAP kinase in articular chondrocytes. J. Biol. Chem., 2008, 283: 1043-1051.

[112]

Fichter M. et al.. Collagen degradation products modulate matrix metalloproteinase expression in cultured articular chondrocytes. J. Orthop. Res., 2006, 24: 63-70.

[113]

Tchetina EV. et al.. Chondrocyte hypertrophy can be induced by a cryptic sequence of type II collagen and is accompanied by the induction of MMP-13 and collagenase activity: implications for development and arthritis. Matrix Biol., 2007, 26: 247-258.

[114]

Yasuda T. Type II collagen peptide stimulates Akt leading to nuclear factor-kappaB activation: its inhibition by hyaluronan. Biomed. Res., 2014, 35: 193-199.

[115]

Lambert C, Borderie D, Dubuc JE, Rannou F, Henrotin Y. Type II collagen peptide Coll2-1 is an actor of synovitis. Osteoarthr. Cartil., 2019, 27: 1680-1691.

[116]

Lees S. et al.. Bioactivity in an aggrecan 32-mer fragment is mediated via toll-like receptor 2. Arthritis Rheumatol., 2015, 67: 1240-1249.

[117]

Babelova A. et al.. Biglycan, a danger signal that activates the NLRP3 inflammasome via toll-like and P2X receptors. J. Biol. Chem., 2009, 284: 24035-24048.

[118]

Schaefer L. et al.. The matrix component biglycan is proinflammatory and signals through Toll-like receptors 4 and 2 in macrophages. J. Clin. Invest., 2005, 115: 2223-2233.

[119]

Hsieh, L. T. et al. Biglycan- and sphingosine kinase-1 signaling crosstalk regulates the synthesis of macrophage chemoattractants. Int. J. Mol. Sci.18, 595 (2017).

[120]

Zeng-Brouwers J, Beckmann J, Nastase MV, Iozzo RV, Schaefer L. De novo expression of circulating biglycan evokes an innate inflammatory tissue response via MyD88/TRIF pathways. Matrix Biol., 2014, 35: 132-142.

[121]

Merline R. et al.. Signaling by the matrix proteoglycan decorin controls inflammation and cancer through PDCD4 and MicroRNA-21. Sci. Signal, 2011, 4: ra75.

[122]

Wang W. et al.. Ligation of TLR2 by versican: a link between inflammation and metastasis. Arch. Med. Res., 2009, 40: 321-323.

[123]

Tang M. et al.. Toll-like receptor 2 activation promotes tumor dendritic cell dysfunction by regulating IL-6 and IL-10 receptor signaling. Cell Rep., 2015, 13: 2851-2864.

[124]

Li D, Wu M. Pattern recognition receptors in health and diseases. Signal Transduct. Target Ther., 2021, 6: 291.

[125]

Song C. et al.. An in-depth analysis of the immunomodulatory mechanisms of intervertebral disc degeneration. JOR Spine, 2022, 5. e1233

[126]

Suresh R, Mosser DM. Pattern recognition receptors in innate immunity, host defense, and immunopathology. Adv. Physiol. Educ., 2013, 37: 284-291.

[127]

Feng P. et al.. Immune exposure: how macrophages interact with the nucleus pulposus. Front. Immunol., 2023, 14. 1155746

[128]

Krock E. et al.. Interleukin-8 as a therapeutic target for chronic low back pain: Upregulation in human cerebrospinal fluid and pre-clinical validation with chronic reparixin in the SPARC-null mouse model. EBioMedicine, 2019, 43: 487-500.

[129]

Rand N, Reichert F, Floman Y, Rotshenker S. Murine nucleus pulposus-derived cells secrete interleukins-1-beta, -6, and -10 and granulocyte-macrophage colony-stimulating factor in cell culture. Spine (Philos. PA 1976), 1997, 22: 2598-2601. 2

[130]

Burke JG. et al.. Human nucleus pulposis can respond to a pro-inflammatory stimulus. Spine (Philos. PA 1976), 2003, 28: 2685-2693.

[131]

Burke JG. et al.. Spontaneous production of monocyte chemoattractant protein-1 and interleukin-8 by the human lumbar intervertebral disc. Spine (Philos. Pa 1976), 2002, 27: 1402-1407.

[132]

Gabr MA. et al.. Interleukin-17 synergizes with IFNgamma or TNFalpha to promote inflammatory mediator release and intercellular adhesion molecule-1 (ICAM-1) expression in human intervertebral disc cells. J. Orthop. Res, 2011, 29: 1-7.

[133]

Wang SS. et al.. IL-17A enhances ADAMTS-7 expression through regulation of TNF-alpha in human nucleus pulposus cells. J. Mol. Histol., 2015, 46: 475-483.

[134]

Cuellar JM. et al.. Cytokine evaluation in individuals with low back pain using discographic lavage. Spine J., 2010, 10: 212-218.

[135]

Liu T, Zhang L, Joo D, Sun SC. NF-kappaB signaling in inflammation. Signal Transduct. Target Ther., 2017, 2: 17023.

[136]

Ahmed, H. et al. Role of adaptor protein myeloid differentiation 88 (MyD88) in post-subarachnoid hemorrhage inflammation: a systematic review. Int. J. Mol. Sci.22, 4185 (2021).

[137]

Xu YR, Lei CQ. TAK1-TABs complex: a central signalosome in inflammatory responses. Front. Immunol., 2020, 11. 608976

[138]

Glaeser JD. et al.. NF-kappaB inhibitor, NEMO-binding domain peptide attenuates intervertebral disc degeneration. Spine J., 2020, 20: 1480-1491.

[139]

Chao-Yang G, Peng C, Hai-Hong Z. Roles of NLRP3 inflammasome in intervertebral disc degeneration. Osteoarthr. Cartil., 2021, 29: 793-801.

[140]

Sun X. et al.. The NLRP3 Inflammasome and Its Role in T1DM. Front. Immunol., 2020, 11: 1595.

[141]

Zarezadeh Mehrabadi A. et al.. The roles of interleukin-1 receptor accessory protein in certain inflammatory conditions. Immunology, 2022, 166: 38-46.

[142]

Fan H. et al.. Necroptosis of nucleus pulposus cells involved in intervertebral disc degeneration through MyD88 signaling. Front. Endocrinol. (Lausanne), 2022, 13. 994307

[143]

Lambert C. et al.. The damage-associated molecular patterns (DAMPs) as potential targets to treat osteoarthritis: perspectives from a review of the literature. Front. Med. (Lausanne), 2020, 7. 607186

[144]

Na HSL, Haneef X, Liu K. W. Old and new damage-associated molecular patterns (DAMPs) in autoimmune diseases. Rheumatol. Autoimmun., 2022, 2: 185-197.

[145]

Yu H, Lin L, Zhang Z, Zhang H, Hu H. Targeting NF-kappaB pathway for the therapy of diseases: mechanism and clinical study. Signal Transduct. Target Ther., 2020, 5: 209.

[146]

Ahmed, A. S. et al. NF-kappaB-associated pain-related neuropeptide expression in patients with degenerative disc disease. Int. J. Mol. Sci.20, 658 (2019).

[147]

Guo Q. et al.. NF-kappaB in biology and targeted therapy: new insights and translational implications. Signal Transduct. Target Ther., 2024, 9: 53.

[148]

Garcia-Garcia, V. A., Alameda, J. P., Page, A. & Casanova, M. L. Role of NF-kappaB in ageing and age-related diseases: lessons from genetically modified mouse models. Cells10, 1906 (2021).

[149]

Zhang GZ. et al.. NF-kappaB signalling pathways in nucleus pulposus cell function and intervertebral disc degeneration. Cell Prolif., 2021, 54. e13057

[150]

David G, Ciurea AV, Iencean SM, Mohan A. Angiogenesis in the degeneration of the lumbar intervertebral disc. J. Med. Life, 2010, 3: 154-161

[151]

Koroth, J. et al. Macrophages and intervertebral disc degeneration. Int. J. Mol. Sci.24, 1367 (2023).

[152]

Li, Y. et al. The potential role and trend of HIF‑1alpha in intervertebral disc degeneration: friend or foe? (Review). Mol. Med. Rep.23, 239 (2021).

[153]

Yamakawa M. et al.. Hypoxia-inducible factor-1 mediates activation of cultured vascular endothelial cells by inducing multiple angiogenic factors. Circ. Res., 2003, 93: 664-673.

[154]

Kwon WK, Moon HJ, Kwon TH, Park YK, Kim JH. The role of hypoxia in angiogenesis and extracellular matrix regulation of intervertebral disc cells during inflammatory reactions. Neurosurgery, 2017, 81: 867-875.

[155]

Zhang H. et al.. SDF1/CXCR4 axis facilitates the angiogenesis via activating the PI3K/AKT pathway in degenerated discs. Mol. Med. Rep., 2020, 22: 4163-4172

[156]

Yang H. et al.. Synergistic effect of VEGF and SDF-1alpha in endothelial progenitor cells and vascular smooth muscle cells. Front. Pharm., 2022, 13: 914347.

[157]

Giraudo E. et al.. Tumor necrosis factor-alpha regulates expression of vascular endothelial growth factor receptor-2 and of its co-receptor neuropilin-1 in human vascular endothelial cells. J. Biol. Chem., 1998, 273: 22128-22135.

[158]

Freemont AJ. et al.. Nerve ingrowth into diseased intervertebral disc in chronic back pain. Lancet, 1997, 350: 178-181.

[159]

Binch AL. et al.. Nerves are more abundant than blood vessels in the degenerate human intervertebral disc. Arthritis Res. Ther., 2015, 17: 370.

[160]

Ashton IK, Roberts S, Jaffray DC, Polak JM, Eisenstein SM. Neuropeptides in the human intervertebral disc. J. Orthop. Res., 1994, 12: 186-192.

[161]

He M. et al.. Overexpression of TIMP3 inhibits discogenic pain by suppressing angiogenesis and the expression of substance P in nucleus pulposus. Mol. Med. Rep., 2020, 21: 1163-1171

[162]

Landreth, G. in Basic Neurochemistry: Molecular, Cellular and Medical Aspects (Lippincott Williams & Wilkins, 1999).

[163]

Iannone F. et al.. Increased expression of nerve growth factor (NGF) and high affinity NGF receptor (p140 TrkA) in human osteoarthritic chondrocytes. Rheumatol. (Oxf.), 2002, 41: 1413-1418.

[164]

Rihl M. et al.. Involvement of neurotrophins and their receptors in spondyloarthritis synovitis: relation to inflammation and response to treatment. Ann. Rheum. Dis., 2005, 64: 1542-1549.

[165]

Purmessur D, Freemont AJ, Hoyland JA. Expression and regulation of neurotrophins in the nondegenerate and degenerate human intervertebral disc. Arthritis Res Ther., 2008, 10. R99

[166]

Binch AL. et al.. Expression and regulation of neurotrophic and angiogenic factors during human intervertebral disc degeneration. Arthritis Res. Ther., 2014, 16: 416.

[167]

Garcia-Cosamalon J. et al.. Intervertebral disc, sensory nerves and neurotrophins: who is who in discogenic pain?. J. Anat., 2010, 217: 1-15.

[168]

Lee FS, Chao MV. Activation of Trk neurotrophin receptors in the absence of neurotrophins. Proc. Natl. Acad. Sci. USA, 2001, 98: 3555-3560.

[169]

Huang EJ, Reichardt LF. Neurotrophins: roles in neuronal development and function. Annu Rev. Neurosci., 2001, 24: 677-736.

[170]

Bibel M, Barde YA. Neurotrophins: key regulators of cell fate and cell shape in the vertebrate nervous system. Genes Dev., 2000, 14: 2919-2937.

[171]

Kruttgen A, Saxena S, Evangelopoulos ME, Weis J. Neurotrophins and neurodegenerative diseases: receptors stuck in traffic?. J. Neuropathol. Exp. Neurol., 2003, 62: 340-350.

[172]

Minnone, G., De Benedetti, F. & Bracci-Laudiero, L. NGF and its receptors in the regulation of inflammatory response. Int. J. Mol. Sci.18, 1028 (2017).

[173]

Covaceuszach S. et al.. The conundrum of the high-affinity NGF binding site formation unveiled?. Biophys. J., 2015, 108: 687-697.

[174]

McGregor CE, English AW. The role of BDNF in peripheral nerve regeneration: activity-dependent treatments and val66Met. Front. Cell Neurosci., 2018, 12: 522.

[175]

Reichardt LF. Neurotrophin-regulated signalling pathways. Philos. Trans. R. Soc. Lond. B Biol. Sci., 2006, 361: 1545-1564.

[176]

Barker PA. p75NTR: a study in contrasts. Cell Death Differ., 1998, 5: 346-356.

[177]

Sajanti A. et al.. A comprehensive p75 neurotrophin receptor gene network and pathway analyses identifying new target genes. Sci. Rep., 2020, 10. 14984

[178]

Chen Y. et al.. Multiple roles of the p75 neurotrophin receptor in the nervous system. J. Int. Med. Res., 2009, 37: 281-288.

[179]

Ceni C. et al.. The p75NTR intracellular domain generated by neurotrophin-induced receptor cleavage potentiates Trk signaling. J. Cell Sci., 2010, 123: 2299-2307.

[180]

Hoyle GW, Mercer EH, Palmiter RD, Brinster RL. Expression of NGF in sympathetic neurons leads to excessive axon outgrowth from ganglia but decreased terminal innervation within tissues. Neuron, 1993, 10: 1019-1034.

[181]

Aloe L, Rocco ML, Bianchi P, Manni L. Nerve growth factor: from the early discoveries to the potential clinical use. J. Transl. Med., 2012, 10. 239

[182]

Bracci-Laudiero, L. M., L. NGF and Immune Regulation. 1849–1876 (Springer, 2014).

[183]

Richardson SM. et al.. Degenerate human nucleus pulposus cells promote neurite outgrowth in neural cells. PLoS One, 2012, 7: e47735.

[184]

Davis BM, Goodness TP, Soria A, Albers KM. Over-expression of NGF in skin causes formation of novel sympathetic projections to trkA-positive sensory neurons. Neuroreport, 1998, 9: 1103-1107.

[185]

Bracci-Laudiero L, De Stefano ME. NGF in early embryogenesis, differentiation, and pathology in the nervous and immune systems. Curr. Top. Behav. Neurosci., 2016, 29: 125-152.

[186]

Causing CG. et al.. Synaptic innervation density is regulated by neuron-derived BDNF. Neuron, 1997, 18: 257-267.

[187]

Benito-Gutierrez E, Garcia-Fernandez J, Comella JX. Origin and evolution of the Trk family of neurotrophic receptors. Mol. Cell Neurosci., 2006, 31: 179-192.

[188]

Triaca, V. et al. hNGF peptides elicit the NGF-TrkA signalling pathway in cholinergic neurons and retain full neurotrophic activity in the DRG assay. Biomolecules10, 216 (2020).

[189]

Zha K. et al.. Nerve growth factor (NGF) and NGF receptors in mesenchymal stem/stromal cells: Impact on potential therapies. Stem Cells Transl. Med., 2021, 10: 1008-1020.

[190]

Daragmeh J, Barriah W, Saad B, Zaid H. Analysis of PI3K pathway components in human cancers. Oncol. Lett., 2016, 11: 2913-2918.

[191]

Molloy NH, Read DE, Gorman AM. Nerve growth factor in cancer cell death and survival. Cancers, 2011, 3: 510-530.

[192]

Mantyh PW, Koltzenburg M, Mendell LM, Tive L, Shelton DL. Antagonism of nerve growth factor-TrkA signaling and the relief of pain. Anesthesiology, 2011, 115: 189-204.

[193]

Turney SG. et al.. Nerve growth factor stimulates axon outgrowth through negative regulation of growth cone actomyosin restraint of microtubule advance. Mol. Biol. Cell, 2016, 27: 500-517.

[194]

Barker PA, Mantyh P, Arendt-Nielsen L, Viktrup L, Tive L. Nerve growth factor signaling and its contribution to pain. J. Pain. Res., 2020, 13: 1223-1241.

[195]

Freemont AJ. et al.. Nerve growth factor expression and innervation of the painful intervertebral disc. J. Pathol., 2002, 197: 286-292.

[196]

Jin, W. Regulation of BDNF-TrkB signaling and potential therapeutic strategies for parkinson’s disease. J. Clin. Med.9, 257 (2020).

[197]

Esvald EE. et al.. CREB family transcription factors are major mediators of BDNF transcriptional autoregulation in cortical neurons. J. Neurosci., 2020, 40: 1405-1426.

[198]

Gupta VK, You Y, Gupta VB, Klistorner A, Graham SL. TrkB receptor signalling: implications in neurodegenerative, psychiatric and proliferative disorders. Int. J. Mol. Sci., 2013, 14: 10122-10142.

[199]

Bonni A. et al.. Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms. Science, 1999, 286: 1358-1362.

[200]

Moya-Alvarado, G. et al. BDNF/TrkB signaling endosomes in axons coordinate CREB/mTOR activation and protein synthesis in the cell body to induce dendritic growth in cortical neurons. Elife12, e77455 (2023).

[201]

Vega JA, Garcia-Suarez O, Hannestad J, Perez-Perez M, Germana A. Neurotrophins and the immune system. J. Anat., 2003, 203: 1-19.

[202]

Krock E. et al.. Nerve growth factor is regulated by Toll-Like receptor 2 in human intervertebral discs. J. Biol. Chem., 2016, 291: 3541-3551.

[203]

Abe Y. et al.. Proinflammatory cytokines stimulate the expression of nerve growth factor by human intervertebral disc cells. Spine (Philos. Pa 1976), 2007, 32: 635-642.

[204]

Airaksinen MS, Saarma M. The GDNF family: signalling, biological functions and therapeutic value. Nat. Rev. Neurosci., 2002, 3: 383-394.

[205]

Saarma, M. in Encyclopedia of Neuroscience (2009).

[206]

Kotliarova A, Sidorova YA. Glial cell line-derived neurotrophic factor family ligands, players at the interface of neuroinflammation and neuroprotection: focus onto the glia. Front. Cell Neurosci., 2021, 15. 679034

[207]

Yamada J. et al.. Expression of glial cell line-derived neurotrophic factor in the human intervertebral disc. Spine (Philos. Pa 1976), 2020, 45: E768-E775.

[208]

Iwasaki, T. et al. Expression of glial-cell-line-derived neurotrophic factor family ligands in human intervertebral discs. Int. J. Mol. Sci.24, 15874 (2023).

[209]

Burbach JP. What are neuropeptides?. Methods Mol. Biol., 2011, 789: 1-36.

[210]

Kepler CK. et al.. Substance P stimulates production of inflammatory cytokines in human disc cells. Spine (Philos. Pa 1976), 2013, 38: E1291-E1299.

[211]

Suvas S. Role of substance P neuropeptide in inflammation, wound healing, and tissue homeostasis. J. Immunol., 2017, 199: 1543-1552.

[212]

Zheng J. et al.. Reactive oxygen species mediate low back pain by upregulating substance P in intervertebral disc degeneration. Oxid. Med. Cell Longev., 2021, 2021. 6681815

[213]

Johnson MB, Young AD, Marriott I. The therapeutic potential of targeting substance P/NK-1R interactions in inflammatory CNS disorders. Front. Cell Neurosci., 2016, 10: 296

[214]

Freidin M, Kessler JA. Cytokine regulation of substance P expression in sympathetic neurons. Proc. Natl. Acad. Sci. USA, 1991, 88: 3200-3203.

[215]

Russell FA, King R, Smillie SJ, Kodji X, Brain SD. Calcitonin gene-related peptide: physiology and pathophysiology. Physiol. Rev., 2014, 94: 1099-1142.

[216]

Kim YJ, Granstein RD. Roles of calcitonin gene-related peptide in the skin, and other physiological and pathophysiological functions. Brain Behav. Immun. Health, 2021, 18. 100361

[217]

Solway B, Bose SC, Corder G, Donahue RR, Taylor BK. Tonic inhibition of chronic pain by neuropeptide Y. Proc. Natl. Acad. Sci. USA, 2011, 108: 7224-7229.

[218]

Dombrowski ME. et al.. Rabbit annulus fibrosus cells express neuropeptide Y, which is influenced by mechanical and inflammatory stress. Neurospine, 2020, 17: 69-76.

[219]

Sun K. et al.. Neuropeptide Y prevents nucleus pulposus cells from cell apoptosis and IL‑1beta‑induced extracellular matrix degradation. Cell Cycle, 2021, 20: 960-977.

[220]

Kameda, T. et al. Expression and activity of TRPA1 and TRPV1 in the intervertebral disc: association with inflammation and matrix remodeling. Int. J. Mol. Sci.20, 1767 (2019).

[221]

Sadowska A. et al.. Differential regulation of TRP channel gene and protein expression by intervertebral disc degeneration and back pain. Sci. Rep., 2019, 9. 18889

[222]

Easson GWD, Savadipour A, Gonzalez C, Guilak F, Tang SY. TRPV4 differentially controls inflammatory cytokine networks during static and dynamic compression of the intervertebral disc. JOR Spine, 2023, 6. e1282

[223]

Easson GWD. et al.. Modulation of TRPV4 protects against degeneration induced by sustained loading and promotes matrix synthesis in the intervertebral disc. FASEB J., 2023, 37. e22714

[224]

Zheng B. et al.. Netrin-1 mediates nerve innervation and angiogenesis leading to discogenic pain. J. Orthop. Transl., 2023, 39: 21-33

[225]

Krames ES. The role of the dorsal root ganglion in the development of neuropathic pain. Pain. Med., 2014, 15: 1669-1685.

[226]

Diwan AD, Melrose J. Intervertebral disc degeneration and how it leads to low back pain. JOR Spine, 2023, 6. e1231

[227]

Li W. et al.. Peripheral and central pathological mechanisms of chronic low back Pain: a narrative review. J. Pain. Res., 2021, 14: 1483-1494.

[228]

Scholz J, Woolf CJ. The neuropathic pain triad: neurons, immune cells and glia. Nat. Neurosci., 2007, 10: 1361-1368.

[229]

Poillot P, Snuggs JW, Le Maitre CL, Huyghe JM. L-type voltage-gated calcium channels partly mediate Mechanotransduction in the intervertebral disc. JOR Spine, 2022, 5. e1213

[230]

Vergne-Salle P, Bertin P. Chronic pain and neuroinflammation. Jt. Bone Spine, 2021, 88. 105222

[231]

Murata Y, Olmarker K, Takahashi I, Takahashi K, Rydevik B. Effects of lumbar sympathectomy on pain behavioral changes caused by nucleus pulposus-induced spinal nerve damage in rats. Eur. Spine J., 2006, 15: 634-640.

[232]

Herrero JF, Laird JM, Lopez-Garcia JA. Wind-up of spinal cord neurones and pain sensation: much ado about something?. Prog. Neurobiol., 2000, 61: 169-203.

[233]

Latremoliere A, Woolf CJ. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J. Pain., 2009, 10: 895-926.

[234]

Cairns BE, Arendt-Nielsen L, Sacerdote P. Perspectives in pain research 2014: neuroinflammation and glial cell activation: the cause of transition from acute to chronic pain?. Scand. J. Pain., 2015, 6: 3-6.

[235]

Lai, A. et al. Annulus fibrosus injury induces acute neuroinflammation and chronic glial response in dorsal root ganglion and spinal cord-an in vivo rat discogenic pain model. Int. J. Mol. Sci.25, 1762 (2024).

[236]

Gruber HE, Hanley ENJr. Human disc cells in monolayer vs 3D culture: cell shape, division and matrix formation. BMC Musculoskelet. Disord., 2000, 1. 1

[237]

Wang JY, Baer AE, Kraus VB, Setton LA. Intervertebral disc cells exhibit differences in gene expression in alginate and monolayer culture. Spine (Philos. Pa 1976), 2001, 26: 1747-1751.

[238]

Horner HA. et al.. Cells from different regions of the intervertebral disc: effect of culture system on matrix expression and cell phenotype. Spine (Philos. Pa 1976), 2002, 27: 1018-1028.

[239]

Chou AI, Reza AT, Nicoll SB. Distinct intervertebral disc cell populations adopt similar phenotypes in three-dimensional culture. Tissue Eng. Part A, 2008, 14: 2079-2087.

[240]

Chen YF. et al.. Insights into the hallmarks of human nucleus pulposus cells with particular reference to cell viability, phagocytic potential and long process formation. Int. J. Med. Sci., 2013, 10: 1805-1816.

[241]

Dai J. et al.. Microfluidic disc-on-a-chip device for mouse intervertebral disc-pitching a next-generation research platform to study disc degeneration. ACS Biomater. Sci. Eng., 2019, 5: 2041-2051.

[242]

Owen PJ, Hangai M, Kaneoka K, Rantalainen T, Belavy DL. Mechanical loading influences the lumbar intervertebral disc. A cross-sectional study in 308 athletes and 71 controls. J. Orthop. Res., 2021, 39: 989-997.

[243]

Xiang P, Luo ZP, Che YJ. Insights into the mechanical microenvironment within the cartilaginous endplate: an emerging role in maintaining disc homeostasis and normal function. Heliyon, 2024, 10. e31162

[244]

McKinley JP. et al.. Design of a flexing organ-chip to model in situ loading of the intervertebral disc. Biomicrofluidics, 2022, 16. 054111

[245]

Xie, W. et al. Intervertebral disc-on-a-chipMF: a new model for mouse disc culture via integrating mechanical loading and dynamic media flow. Adv. Mater.Technol.8, 2300606 (2023).

[246]

Son HG. et al.. Intervertebral disc organ-on-a-chip: an innovative model to study monocyte extravasation during nucleus pulposus degeneration. Lab Chip, 2023, 23: 2819-2828.

[247]

Hwang MH. et al.. Spine-on-a-chip: human annulus fibrosus degeneration model for simulating the severity of intervertebral disc degeneration. Biomicrofluidics, 2017, 11. 064107

[248]

Hwang MH, Son HG, Kim J, Choi H. In vitro model of distinct catabolic and inflammatory response patterns of endothelial cells to intervertebral disc cell degeneration. Sci. Rep., 2020, 10. 20596

[249]

Kague E. et al.. 3D assessment of intervertebral disc degeneration in zebrafish identifies changes in bone density that prime disc disease. Bone Res., 2021, 9: 39.

[250]

Chengguo SU. et al.. Effect of Tuina along “bladder meridian” alleviating intervertebral disc degeneration by regulating the transforming growth factor-beta1/Smad signaling pathway in a rabbit model. J. Tradit. Chin. Med., 2023, 43: 991-1000

[251]

Yang K. et al.. Comparisons between needle puncture and chondroitinase ABC to induce intervertebral disc degeneration in rabbits. Eur. Spine J., 2022, 31: 2788-2800.

[252]

Zhang Y, Drapeau S, Howard SA, Thonar EJ, Anderson DG. Transplantation of goat bone marrow stromal cells to the degenerating intervertebral disc in a goat disc injury model. Spine (Philos. Pa 1976), 2011, 36: 372-377.

[253]

Peredo AP. et al.. Tension-activated nanofiber patches delivering an anti-inflammatory drug improve repair in a goat intervertebral disc herniation model. Sci. Transl. Med., 2023, 15: eadf1690.

[254]

Zhang Y. et al.. Histological features of the degenerating intervertebral disc in a goat disc-injury model. Spine (Philos. Pa 1976), 2011, 36: 1519-1527.

[255]

Yuan, Q. et al. Autologous mesenchymal stromal cells combined with gelatin sponge for repair intervertebral disc defect after discectomy: a preclinical study in a goat model. Front. Biosci. (Landmark Ed. 27, 131 (2022) .

[256]

Woiciechowsky C. et al.. Regeneration of nucleus pulposus tissue in an ovine intervertebral disc degeneration model by cell-free resorbable polymer scaffolds. J. Tissue Eng. Regen. Med., 2014, 8: 811-820.

[257]

Vadala G. et al.. Novel stepwise model of intervertebral disc degeneration with intact annulus fibrosus to test regeneration strategies. J. Orthop. Res., 2018, 36: 2460-2468.

[258]

Daly CD. et al.. A comparison of two ovine lumbar intervertebral disc injury models for the evaluation and development of novel regenerative therapies. Glob. Spine J., 2018, 8: 847-859.

[259]

Oehme D. et al.. Mesenchymal progenitor cells combined with pentosan polysulfate mediating disc regeneration at the time of microdiscectomy: a preliminary study in an ovine model. J. Neurosurg. Spine, 2014, 20: 657-669.

[260]

Melrose J. et al.. Mechanical destabilization induced by controlled annular incision of the intervertebral disc dysregulates metalloproteinase expression and induces disc degeneration. Spine (Philos. Pa 1976), 2012, 37: 18-25.

[261]

Fuller ES, Shu C, Smith MM, Little CB, Melrose J. Hyaluronan oligosaccharides stimulate matrix metalloproteinase and anabolic gene expression in vitro by intervertebral disc cells and annular repair in vivo. J. Tissue Eng. Regen. Med, 2018, 12: e216-e226.

[262]

Shu CC. et al.. Efficacy of administered mesenchymal stem cells in the initiation and co-ordination of repair processes by resident disc cells in an ovine (Ovis aries) large destabilizing lesion model of experimental disc degeneration. JOR Spine, 2018, 1. e1037

[263]

Willems N. et al.. Inflammatory profiles in canine intervertebral disc degeneration. BMC Vet. Res, 2016, 12: 10.

[264]

Tellegen AR. et al.. Intradiscal application of a PCLA-PEG-PCLA hydrogel loaded with celecoxib for the treatment of back pain in canines: what’s in it for humans?. J. Tissue Eng. Regen. Med., 2018, 12: 642-652.

[265]

Tellegen AR. et al.. Intradiscal delivery of celecoxib-loaded microspheres restores intervertebral disc integrity in a preclinical canine model. J. Control Release, 2018, 286: 439-450.

[266]

Rudnik-Jansen I. et al.. Safety of intradiscal delivery of triamcinolone acetonide by a poly(esteramide) microsphere platform in a large animal model of intervertebral disc degeneration. Spine J., 2019, 19: 905-919.

[267]

Platenberg RC, Hubbard GB, Ehler WJ, Hixson CJ. Spontaneous disc degeneration in the baboon model: magnetic resonance imaging and histopathologic correlation. J. Med. Primatol., 2001, 30: 268-272.

[268]

Nuckley DJ. et al.. Intervertebral disc degeneration in a naturally occurring primate model: radiographic and biomechanical evidence. J. Orthop. Res., 2008, 26: 1283-1288.

[269]

Wang J. et al.. Correlation between motor behavior and age-related intervertebral disc degeneration in cynomolgus monkeys. JOR Spine, 2022, 5: e1183.

[270]

Shi C. et al.. Development of an in vivo mouse model of discogenic low back pain. J. Cell Physiol., 2018, 233: 6589-6602.

[271]

Yang G, Chen L, Gao Z, Wang Y. Implication of microglia activation and CSF-1/CSF-1Rpathway in lumbar disc degeneration-related back pain. Mol. Pain., 2018, 14. 1744806918811238

[272]

Wawrose RA. et al.. Percutaneous lumbar annular puncture: a rat model to study intervertebral disc degeneration and pain-related behavior. JOR Spine, 2022, 5: e1202.

[273]

Glaeser JD. et al.. Optimization of a rat lumbar IVD degeneration model for low back pain. JOR Spine, 2020, 3. e1092

[274]

Olmarker K. Puncture of a lumbar intervertebral disc induces changes in spontaneous pain behavior: an experimental study in rats. Spine (Philos. Pa 1976), 2008, 33: 850-855.

[275]

Barbe MF. et al.. Characterization of pain-related behaviors in a rat model of acute-to-chronic low back pain: single vs. multi-level disc injury. Front Pain. Res. (Lausanne), 2024, 5. 1394017

[276]

Lee S, Millecamps M, Foster DZ, Stone LS. Long-term histological analysis of innervation and macrophage infiltration in a mouse model of intervertebral disc injury-induced low back pain. J. Orthop. Res., 2020, 38: 1238-1247.

[277]

Millecamps M, Lee S, Foster DZ, Stone LS. Disc degeneration spreads: long-term behavioural, histologic and radiologic consequences of a single-level disc injury in active and sedentary mice. Eur. Spine J., 2021, 30: 2238-2246.

[278]

Nojima D. et al.. Efficacy of anti-NaV1.7 antibody on the sensory nervous system in a rat model of lumbar intervertebral disc injury. Yonsei Med. J., 2016, 57: 748-753.

[279]

Miyagi M. et al.. Disk injury in rats produces persistent increases in pain-related neuropeptides in dorsal root ganglia and spinal cord glia but only transient increases in inflammatory mediators: pathomechanism of chronic diskogenic low back pain. Spine (Philos. Pa 1976), 2011, 36: 2260-2266.

[280]

Chen X. et al.. Reactive oxygen species induced upregulation of TRPV1 in dorsal root ganglia results in low back pain in rats. J. Inflamm. Res., 2024, 17: 2245-2256.

[281]

Sadamasu A. et al.. Upregulation of NaV1.7 in dorsal root ganglia after intervertebral disc injury in rats. Spine (Philos. Pa 1976), 2014, 39: E421-E426.

[282]

Li Y. et al.. Activation of satellite cells in the dorsal root ganglia in a disc-punctured rat model. J. Orthop. Sci., 2011, 16: 433-438.

[283]

Kim JS. et al.. The rat intervertebral disk degeneration pain model: relationships between biological and structural alterations and pain. Arthritis Res. Ther., 2011, 13. R165

[284]

Lai A. et al.. Assessment of functional and behavioral changes sensitive to painful disc degeneration. J. Orthop. Res., 2015, 33: 755-764.

[285]

Miyagi M. et al.. Assessment of pain behavior in a rat model of intervertebral disc injury using the CatWalk gait analysis system. Spine (Philos. Pa 1976), 2013, 38: 1459-1465.

[286]

Suzuki, H. et al. Injection of ultra-purified stem cells with sodium alginate reduces discogenic pain in a rat model. Cells12, 505 (2023).

[287]

Zhang H, La Marca F, Hollister SJ, Goldstein SA, Lin CY. Developing consistently reproducible intervertebral disc degeneration at rat caudal spine by using needle puncture. J. Neurosurg. Spine, 2009, 10: 522-530.

[288]

Liao Z. et al.. Exosomes from mesenchymal stem cells modulate endoplasmic reticulum stress to protect against nucleus pulposus cell death and ameliorate intervertebral disc degeneration in vivo. Theranostics, 2019, 9: 4084-4100.

[289]

Chen C. et al.. Autologous fibroblasts induce fibrosis of the nucleus pulposus to maintain the stability of degenerative intervertebral discs. Bone Res., 2020, 8: 7.

[290]

Martin JT. et al.. Needle puncture injury causes acute and long-term mechanical deficiency in a mouse model of intervertebral disc degeneration. J. Orthop. Res., 2013, 31: 1276-1282.

[291]

Ohnishi T. et al.. In vivo mouse intervertebral disc degeneration model based on a new histological classification. PLoS One, 2016, 11: e0160486.

[292]

Au TYK. et al.. Transformation of resident notochord-descendent nucleus pulposus cells in mouse injury-induced fibrotic intervertebral discs. Aging Cell, 2020, 19. e13254

[293]

Millecamps M, Stone LS. Delayed onset of persistent discogenic axial and radiating pain after a single-level lumbar intervertebral disc injury in mice. Pain, 2018, 159: 1843-1855.

[294]

Lee M. et al.. Complete Freund’s adjuvant-induced intervertebral discitis as an animal model for discogenic low back pain. Anesth. Analg., 2009, 109: 1287-1296.

[295]

Sasaki N, Kikuchi S, Konno S, Sekiguchi M, Watanabe K. Anti-TNF-alpha antibody reduces pain-behavioral changes induced by epidural application of nucleus pulposus in a rat model depending on the timing of administration. Spine (Philos. PA 1976), 2007, 32: 413-416.

[296]

Yang L. et al.. Effective modulation of inflammation and oxidative stress for enhanced regeneration of intervertebral discs using 3D porous hybrid protein nanoscaffold. Adv. Mater., 2023, 35. e2303021

[297]

Ishiguro H. et al.. Intervertebral disc regeneration with an adipose mesenchymal stem cell-derived tissue-engineered construct in a rat nucleotomy model. Acta Biomater., 2019, 87: 118-129.

[298]

Moon CS, Lim TH, Hong J, Sul D, Kim N. Assessment of a discogenic pain animal model induced by applying continuous shear force to intervertebral discs. Pain. Physician, 2023, 26: E181-E189.

[299]

Xie W. et al.. A mouse coccygeal intervertebral disc degeneration model with tail-looping constructed using a suturing method. Anim. Model Exp. Med., 2025.

[300]

Matyas JR, Gutmann A, Randev J, Hurtig M, Bertram JE. Intra-articular anaesthesia mitigates established pain in experimental osteoarthritis: a preliminary study of gait impulse redistribution as a biomarker of analgesia pharmacodynamics. Osteoarthr. Cartil., 2013, 21: 1365-1373.

[301]

Castel D, Willentz E, Doron O, Brenner O, Meilin S. Characterization of a porcine model of post-operative pain. Eur. J. Pain., 2014, 18: 496-505.

[302]

Wu B. et al.. An overview of CEST MRI for non-MR physicists. EJNMMI Phys., 2016, 3. 19

[303]

Melkus G, Grabau M, Karampinos DC, Majumdar S. Ex vivo porcine model to measure pH dependence of chemical exchange saturation transfer effect of glycosaminoglycan in the intervertebral disc. Magn. Reson Med., 2014, 71: 1743-1749.

[304]

Zhou J, Payen JF, Wilson DA, Traystman RJ, van Zijl PC. Using the amide proton signals of intracellular proteins and peptides to detect pH effects in MRI. Nat. Med., 2003, 9: 1085-1090.

[305]

Sun PZ. et al.. Relaxation-compensated fast multislice amide proton transfer (APT) imaging of acute ischemic stroke. Magn. Reson Med., 2008, 59: 1175-1182.

[306]

Sun PZ, Zhou J, Sun W, Huang J, van Zijl PC. Detection of the ischemic penumbra using pH-weighted MRI. J. Cereb. Blood Flow. Metab., 2007, 27: 1129-1136.

[307]

Zhou J, van Zijl PC. Defining an acidosis-based ischemic penumbra from pH-weighted MRI. Transl. Stroke Res., 2011, 3: 76-83.

[308]

Liu Q. et al.. Detection of low back pain using pH level-dependent imaging of the intervertebral disc using the ratio of R1rho dispersion and -OH chemical exchange saturation transfer (RROC). Magn. Reson Med., 2015, 73: 1196-1205.

[309]

Vinogradov E, Sherry AD, Lenkinski RE. CEST: from basic principles to applications, challenges and opportunities. J. Magn. Reson, 2013, 229: 155-172.

[310]

Wu R, Xiao G, Zhou IY, Ran C, Sun PZ. Quantitative chemical exchange saturation transfer (qCEST) MRI - omega plot analysis of RF-spillover-corrected inverse CEST ratio asymmetry for simultaneous determination of labile proton ratio and exchange rate. NMR Biomed., 2015, 28: 376-383.

[311]

Wu R, Longo DL, Aime S, Sun PZ. Quantitative description of radiofrequency (RF) power-based ratiometric chemical exchange saturation transfer (CEST) pH imaging. NMR Biomed., 2015, 28: 555-565.

[312]

Sun PZ, Xiao G, Zhou IY, Guo Y, Wu R. A method for accurate pH mapping with chemical exchange saturation transfer (CEST) MRI. Contrast Media Mol. Imaging, 2016, 11: 195-202.

[313]

Millecamps M, Tajerian M, Naso L, Sage HE, Stone LS. Lumbar intervertebral disc degeneration associated with axial and radiating low back pain in ageing SPARC-null mice. Pain, 2012, 153: 1167-1179.

[314]

Corey SM, Vizzard MA, Bouffard NA, Badger GJ, Langevin HM. Stretching of the back improves gait, mechanical sensitivity and connective tissue inflammation in a rodent model. PLoS One, 2012, 7: e29831.

[315]

Khosravi H. et al.. Back mechanical sensitivity assessment in the rat for mechanistic investigation of chronic back pain. J. Vis. Exp., 2022.

[316]

Lai A. et al.. Annular puncture with tumor necrosis factor-alpha injection enhances painful behavior with disc degeneration in vivo. Spine J., 2016, 16: 420-431.

[317]

Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. Quantitative assessment of tactile allodynia in the rat paw. J. Neurosci. Methods, 1994, 53: 55-63.

[318]

Millecamps M, Czerminski JT, Mathieu AP, Stone LS. Behavioral signs of axial low back pain and motor impairment correlate with the severity of intervertebral disc degeneration in a mouse model. Spine J., 2015, 15: 2524-2537.

[319]

Deuis JR, Dvorakova LS, Vetter I. Methods used to evaluate pain behaviors in rodents. Front. Mol. Neurosci., 2017, 10: 284.

[320]

Randall LO, Selitto JJ. A method for measurement of analgesic activity on inflamed tissue. Arch. Int Pharmacodyn. Ther., 1957, 111: 409-419

[321]

Bigelow LJ, Pope EK, MacDonald DS, Rock JE, Bernard PB. Getting a handle on rat familiarization: the impact of handling protocols on classic tests of stress in Rattus norvegicus. Lab Anim., 2023, 57: 259-269.

[322]

Sensini F. et al.. The impact of handling technique and handling frequency on laboratory mouse welfare is sex-specific. Sci. Rep., 2020, 10. 17281

[323]

Yoon C, Wook YY, Sik NH, Ho KS, Mo CJ. Behavioral signs of ongoing pain and cold allodynia in a rat model of neuropathic pain. Pain, 1994, 59: 369-376.

[324]

Ramabadran K, Bansinath M, Turndorf H, Puig MM. Tail immersion test for the evaluation of a nociceptive reaction in mice. Methodol. Consid. J. Pharm. Methods, 1989, 21: 21-31.

[325]

Brenner DS, Golden JP, Gereau RWT. A novel behavioral assay for measuring cold sensation in mice. PLoS One, 2012, 7: e39765.

[326]

Pizziketti RJ, Pressman NS, Geller EB, Cowan A, Adler MW. Rat cold water tail-flick: a novel analgesic test that distinguishes opioid agonists from mixed agonist-antagonists. Eur. J. Pharm., 1985, 119: 23-29.

[327]

Hargreaves K, Dubner R, Brown F, Flores C, Joris J. A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain, 1988, 32: 77-88.

[328]

Munier JJ. et al.. Simultaneous monitoring of mouse grip strength, force profile, and cumulative force profile distinguishes muscle physiology following surgical, pharmacologic and diet interventions. Sci. Rep., 2022, 12. 16428

[329]

Steru L, Chermat R, Thierry B, Simon P. The tail suspension test: a new method for screening antidepressants in mice. Psychopharmacol. (Berl.), 1985, 85: 367-370.

[330]

Miyagi M. et al.. ISSLS prize winner: disc dynamic compression in rats produces long-lasting increases in inflammatory mediators in discs and induces long-lasting nerve injury and regeneration of the afferent fibers innervating discs: a pathomechanism for chronic discogenic low back pain. Spine (Philos. Pa 1976), 2012, 37: 1810-1818.

[331]

Pattison LA. et al.. Digging deeper into pain: an ethological behavior assay correlating well-being in mice with human pain experience. Pain, 2024, 165: 1761-1773.

[332]

Piel MJ, Kroin JS, van Wijnen AJ, Kc R, Im HJ. Pain assessment in animal models of osteoarthritis. Gene, 2014, 537: 184-188.

[333]

Leimer EM. et al.. Behavioral compensations and neuronal remodeling in a rodent model of chronic intervertebral disc degeneration. Sci. Rep., 2019, 9. 3759

[334]

Kim HT, Uchimoto K, Duellman T, Yang J. Automated assessment of pain in rats using a voluntarily accessed static weight-bearing test. Physiol. Behav., 2015, 151: 139-146.

[335]

Park EH. et al.. Disc degeneration induces a mechano-sensitization of disc afferent nerve fibers that associates with low back pain. Osteoarthr. Cartil., 2019, 27: 1608-1617.

[336]

Xu L. et al.. The anti-NGF antibody muMab 911 both prevents and reverses pain behaviour and subchondral osteoclast numbers in a rat model of osteoarthritis pain. Osteoarthr. Cartil., 2016, 24: 1587-1595.

[337]

Hamers FP, Lankhorst AJ, van Laar TJ, Veldhuis WB, Gispen WH. Automated quantitative gait analysis during overground locomotion in the rat: its application to spinal cord contusion and transection injuries. J. Neurotrauma, 2001, 18: 187-201.

[338]

Fukui D. et al.. Gait abnormality due to spinal instability after lumbar facetectomy in the rat. Eur. Spine J., 2015, 24: 2085-2094.

[339]

Huang Y. et al.. Associations of lumber disc degeneration with paraspinal muscles myosteatosis in discogenic low back pain. Front. Endocrinol. (Lausanne), 2022, 13. 891088

[340]

Urban R, Scherrer G, Goulding EH, Tecott LH, Basbaum AI. Behavioral indices of ongoing pain are largely unchanged in male mice with tissue or nerve injury-induced mechanical hypersensitivity. Pain, 2011, 152: 990-1000.

[341]

Castel D, Sabbag I, Nasaev E, Peng S, Meilin S. Open field and a behavior score in PNT model for neuropathic pain in pigs. J. Pain. Res., 2018, 11: 2279-2293.

[342]

Xin J. et al.. Treatment of intervertebral disc degeneration. Orthop. Surg., 2022, 14: 1271-1280.

[343]

Kuijpers T. et al.. A systematic review on the effectiveness of pharmacological interventions for chronic non-specific low-back pain. Eur. Spine J., 2011, 20: 40-50.

[344]

Jones CMP. et al.. Analgesia for non-specific low back pain. BMJ, 2024, 385: e080064.

[345]

Enthoven WT, Roelofs PD, Deyo RA, van Tulder MW, Koes BW. Non-steroidal anti-inflammatory drugs for chronic low back pain. Cochrane Database Syst. Rev., 2016, 2: CD012087

[346]

Qureshi, O. & Dua, A. COX Inhibitors. in StatPearls [Internet]. (StatPearls Publishing, Treasure Island (FL), 2024). Available from: https://www.ncbi.nlm.nih.gov/books/NBK549795/.

[347]

Smith CJ. et al.. Pharmacological analysis of cyclooxygenase-1 in inflammation. Proc. Natl. Acad. Sci. USA, 1998, 95: 13313-13318.

[348]

Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Arterioscler Thromb. Vasc. Biol., 2011, 31: 986-1000.

[349]

Roelofs PD, Deyo RA, Koes BW, Scholten RJ, van Tulder MW. Non-steroidal anti-inflammatory drugs for low back pain. Cochrane Database Syst. Rev., 2008, 2008: CD000396

[350]

Vonkeman HE, van de Laar MA. Nonsteroidal anti-inflammatory drugs: adverse effects and their prevention. Semin. Arthritis Rheum., 2010, 39: 294-312.

[351]

Pathan H, Williams J. Basic opioid pharmacology: an update. Br. J. Pain., 2012, 6: 11-16.

[352]

Bovill JG. Mechanisms of actions of opioids and non-steroidal anti-inflammatory drugs. Eur. J. Anaesthesiol. Suppl., 1997, 15: 9-15.

[353]

Knapp, C. M. in Encyclopedia of the Human Brain 729-739 (Elsevier, 2002).

[354]

Lambert DG. Opioids and opioid receptors; understanding pharmacological mechanisms as a key to therapeutic advances and mitigation of the misuse crisis. BJA Open, 2023, 6. 100141

[355]

Webster LR. Risk factors for opioid-use disorder and overdose. Anesth. Analg., 2017, 125: 1741-1748.

[356]

Moshfegh J, George SZ, Sun E. Risk and risk factors for chronic opioid use among opioid-naive patients with newly diagnosed musculoskeletal pain in the neck, shoulder, knee, or low back. Ann. Intern Med., 2019, 170: 504-505.

[357]

Carassiti, M. et al. Epidural steroid injections for low back pain: a narrative review. Int. J. Environ. Res. Pub. Health19, 231 (2021).

[358]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT04930211 (2021).

[359]

Vyvey M. Steroids as pain relief adjuvants. Can. Fam. Physician, 2010, 56: 1295-1297

[360]

Katz JN, Zimmerman ZE, Mass H, Makhni MC. Diagnosis and management of lumbar spinal stenosis: a review. JAMA, 2022, 327: 1688-1699.

[361]

Ballard, T. & Chargui, S. in StatPearls (2024).

[362]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT01799616 (2013).

[363]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT03708926 (2018).

[364]

Akel, M., Patel, P. & Parmar, M. Abaloparatide. in StatPearls [Internet]. (StatPearls Publishing. Treasure Island (FL), 2024). Available from: https://www.ncbi.nlm.nih.gov/books/NBK587447/.

[365]

Vandooren J, Itoh Y. Alpha-2-macroglobulin in inflammation, immunity and infections. Front. Immunol., 2021, 12. 803244

[366]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT03307876 (2016).

[367]

Evans L, O’Donohoe T, Morokoff A, Drummond K. The role of spinal surgery in the treatment of low back pain. Med. J. Aust., 2023, 218: 40-45.

[368]

Reid PC, Morr S, Kaiser MG. State of the union: a review of lumbar fusion indications and techniques for degenerative spine disease. J. Neurosurg. Spine, 2019, 31: 1-14.

[369]

Fan H. et al.. Comparison of functional outcome and quality of life in patients with idiopathic scoliosis treated by spinal fusion. Med. (Baltim.), 2016, 95: e3289.

[370]

Kumar MN, Jacquot F, Hall H. Long-term follow-up of functional outcomes and radiographic changes at adjacent levels following lumbar spine fusion for degenerative disc disease. Eur. Spine J., 2001, 10: 309-313.

[371]

Vraa ML, Myers CA, Young JL, Rhon DI. More than 1 in 3 patients with chronic low back pain continue to use opioids long-term after spinal fusion: a systematic review. Clin. J. Pain., 2021, 38: 222-230.

[372]

Zhao L, Manchikanti L, Kaye AD, Abd-Elsayed A. Treatment of discogenic low back pain: current treatment strategies and future options-a literature review. Curr. Pain. Headache Rep., 2019, 23. 86

[373]

Jacobs, W. et al. Total disc replacement for chronic back pain in the presence of disc degeneration. Cochrane Database Syst. Rev. CD008326 (2012). https://doi.org/10.1002/14651858.CD008326.pub2

[374]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT04004156 (2019).

[375]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT03716947 (2018).

[376]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT05508360 (2022).

[377]

Everts, P., Onishi, K., Jayaram, P., Lana, J. F. & Mautner, K. Platelet-rich plasma: new performance understandings and therapeutic considerations in 2020. Int. J. Mol. Sci.21, 7794 (2020).

[378]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT03197415 (2017).

[379]

Dhurat R, Sukesh M. Principles and methods of preparation of platelet-rich plasma: a review and author’s perspective. J. Cutan. Aesthet. Surg., 2014, 7: 189-197.

[380]

Li P, Zhang R, Zhou Q. Efficacy of platelet-rich plasma in retarding intervertebral disc degeneration: a meta-analysis of animal studies. Biomed. Res. Int., 2017, 20177919201

[381]

Soufi, K. H., Castillo, J. A., Rogdriguez, F. Y., DeMesa, C. J. & Ebinu, J. O. Potential role for stem cell regenerative therapy as a treatment for degenerative disc disease and low back pain: a systematic review. Int. J. Mol. Sci.24, 8893 (2023).

[382]

Sakai D, Andersson GB. Stem cell therapy for intervertebral disc regeneration: obstacles and solutions. Nat. Rev. Rheumatol., 2015, 11: 243-256.

[383]

Tong W. et al.. Cell therapy for the degenerating intervertebral disc. Transl. Res., 2017, 181: 49-58.

[384]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT01860417 (2013).

[385]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT04759105 (2021).

[386]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT04530071 (2020).

[387]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT02338271 (2015).

[388]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT05201287 (2021).

[389]

US National Library of Medicine. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT04727385 (2021).

[390]

Gruber HE, Hoelscher GL, Ingram JA, Hanley ENJr. Genome-wide analysis of pain-, nerve- and neurotrophin -related gene expression in the degenerating human annulus. Mol. Pain., 2012, 8. 63

[391]

Sun K. et al.. CGRP regulates nucleus pulposus cell apoptosis and inflammation via the MAPK/NF-kappaB signaling pathways during intervertebral disc degeneration. Oxid. Med. Cell Longev., 2021, 2021. 2958584

[392]

Liu S. et al.. TRPV1 channel activated by the PGE2/EP4 pathway mediates spinal hypersensitivity in a mouse model of vertebral endplate degeneration. Oxid. Med. Cell Longev., 2021, 2021. 9965737

[393]

Peng Y. et al.. Multifunctional annulus fibrosus matrix prevents disc-related pain via inhibiting neuroinflammation and sensitization. Acta Biomater., 2023, 170: 288-302.

[394]

Krock E, Millecamps M, Currie JB, Stone LS, Haglund L. Low back pain and disc degeneration are decreased following chronic toll-like receptor 4 inhibition in a mouse model. Osteoarthr. Cartil., 2018, 26: 1236-1246.

[395]

Miyagi M. et al.. ISSLS Prize winner: increased innervation and sensory nervous system plasticity in a mouse model of low back pain due to intervertebral disc degeneration. Spine (Philos. Pa 1976), 2014, 39: 1345-1354.

[396]

Margarit C. et al.. Genetic contribution in low back pain: a prospective genetic association study. Pain. Pr., 2019, 19: 836-847.

[397]

Guo Z. et al.. The role of IL-6 and TMEM100 in lumbar discogenic pain and the mechanism of the glycine-serine-threonine metabolic axis: a metabolomic and molecular biology study. J. Pain. Res., 2023, 16: 437-461.

[398]

Song XX, Jin LY, Li XF, Luo Y, Yu BW. Substance P mediates estrogen modulation proinflammatory cytokines release in intervertebral disc. Inflammation, 2021, 44: 506-517.

[399]

Aripaka SS, Bech-Azeddine R, Jorgensen LM, Mikkelsen JD. Transient receptor potential (TRP) channels mRNA transcripts in the lumbar intervertebral discs: biomarkers for inflammation, pain, disability, and clinical outcome. Mol. Cell Biochem., 2023, 478: 121-130.

[400]

Hiyama A, Suyama K, Sakai D, Tanaka M, Watanabe M. Correlational analysis of chemokine and inflammatory cytokine expression in the intervertebral disc and blood in patients with lumbar disc disease. J. Orthop. Res., 2022, 40: 1213-1222.

[401]

Lee JM. et al.. Interleukin-1beta induces angiogenesis and innervation in human intervertebral disc degeneration. J. Orthop. Res., 2011, 29: 265-269.

[402]

Pravdyuk, N. G. et al. Immunomorphogenesis in degenerative disc disease: the role of proinflammatory cytokines and angiogenesis factors. Biomedicines11, 2184 (2023).

[403]

Banimostafavi ES. et al.. Determining serum levels of IL-10 and IL-17 in patients with low back pain caused by lumbar disc degeneration. Infect. Disord. Drug Targets, 2021, 21: e270421185135.

[404]

Song XX, Jin LY, Li Q, Li XF, Luo Y. Estrogen receptor beta/substance P signaling in spinal cord mediates antinociceptive effect in a mouse model of discogenic low back pain. Front. Cell Neurosci., 2022, 16: 1071012.

[405]

Noorwali H. et al.. Link N as a therapeutic agent for discogenic pain. JOR Spine, 2018, 1: e1008.

[406]

Tonelli Enrico V. et al.. The association of biomarkers with pain and function in acute and subacute low back pain: a secondary analysis of an RCT. BMC Musculoskelet. Disord., 2022, 23. 1059

[407]

Staszkiewicz R. et al.. Usefulness of detecting brain-derived neurotrophic factor in intervertebral disc degeneration of the lumbosacral spine. Med. Sci. Monit., 2023, 29: e938663

[408]

Aoki Y. et al.. Axonal growth potential of lumbar dorsal root ganglion neurons in an organ culture system: response of nerve growth factor-sensitive neurons to neuronal injury and an inflammatory cytokine. Spine (Philos. Pa 1976), 2007, 32: 857-863.

[409]

Sugiura A. et al.. Existence of nerve growth factor receptors, tyrosine kinase a and p75 neurotrophin receptors in intervertebral discs and on dorsal root ganglion neurons innervating intervertebral discs in rats. Spine (Philos. Pa 1976), 2008, 33: 2047-2051.

[410]

Caparaso, S. M., Sankaranarayanan, I., Lillyman, D. J., Price, T. J. & Wachs, R. A. Single-nuclei RNA sequencing reveals distinct transcriptomic signatures of rat dorsal root ganglia in a chronic discogenic low back pain model. bioRxiv, 2025.2002.2019.639130 (2025). https://doi.org/10.1101/2025.02.19.639130

[411]

Tian S. et al.. Nucleus pulposus cells regulate macrophages in degenerated intervertebral discs via the integrated stress response-mediated CCL2/7-CCR2 signaling pathway. Exp. Mol. Med., 2024, 56: 408-421.

[412]

Li Z, Li Y, Li Z. Low-level miR-199 contribute to neuropathic low back pain via TRPV1 by regulating the production of pro-inflammatory cytokines on macrophage. Turk. Neurosurg., 2024, 34: 299-307

[413]

Li Z, Zhou Y, Li Z. NFKB1 signalling activation contributes to TRPV1 over-expression via repressing MiR-375 and MiR-455: a study on neuropathic low back pain. Folia Biol., 2022, 68: 105-111.

[414]

Rodrigues, P., Ruviaro, N. A. & Trevisan, G. TRPV4 role in neuropathic pain mechanisms in rodents. Antioxidants12, 24 (2022).

[415]

Fozzato S. et al.. TRPV4 and TRPM8 as putative targets for chronic low back pain alleviation. Pflug. Arch., 2021, 473: 151-165.

[416]

Slouma, M. et al. Pro-inflammatory cytokines in patients with low back pain: a comparative study. Reumatol. Clin. (Engl. Ed.19, 244–248 (2023).

[417]

Zhang Y. et al.. Intervertebral disc cells produce interleukins found in patients with back pain. Am. J. Phys. Med. Rehabil., 2016, 95: 407-415.

[418]

Bucknill AT. et al.. Nerve fibers in lumbar spine structures and injured spinal roots express the sensory neuron-specific sodium channels SNS/PN3 and NaN/SNS2. Spine (Philos. Pa 1976), 2002, 27: 135-140.

[419]

Matta, A. et al. A single injection of NTG-101 reduces the expression of pain-related neurotrophins in a canine model of degenerative disc disease. Int. J. Mol. Sci. 23, 5717 (2022).

[420]

Bu G. et al.. Increased expression of netrin-1 and its deleted in colorectal cancer receptor in human diseased lumbar intervertebral disc compared with autopsy control. Spine (Philos. Pa 1976), 2012, 37: 2074-2081.

[421]

Xie W, Strong JA, Ye L, Mao JX, Zhang JM. Knockdown of sodium channel NaV1.6 blocks mechanical pain and abnormal bursting activity of afferent neurons in inflamed sensory ganglia. Pain, 2013, 154: 1170-1180.

[422]

Xie W, Zhang J, Strong JA, Zhang JM. Role of Na(V)1.6 and Na(V)beta4 sodium channel subunits in a rat model of low back pain induced by compression of the dorsal root ganglia. Neuroscience, 2019, 402: 51-65.

[423]

Yan J. et al.. Hyperexcitability and sensitization of sodium channels of dorsal root ganglion neurons in a rat model of lumber disc herniation. Eur. Spine J., 2016, 25: 177-185.

[424]

Chuah YJ. et al.. Scaffold-free tissue engineering with aligned bone marrow stromal cell sheets to recapitulate the microstructural and biochemical composition of annulus fibrosus. Acta Biomater., 2020, 107: 129-137.

[425]

Peng C. et al.. miR-183 cluster scales mechanical pain sensitivity by regulating basal and neuropathic pain genes. Science, 2017, 356: 1168-1171.

[426]

Vo N. et al.. Accelerated aging of intervertebral discs in a mouse model of progeria. J. Orthop. Res., 2010, 28: 1600-1607.

[427]

Choi H. et al.. A novel mouse model of intervertebral disc degeneration shows altered cell fate and matrix homeostasis. Matrix Biol., 2018, 70: 102-122.

[428]

Boyd LM. et al.. Early-onset degeneration of the intervertebral disc and vertebral end plate in mice deficient in type IX collagen. Arthritis Rheum., 2008, 58: 164-171.

[429]

Hasegawa T. et al.. Regenerative effects of platelet-rich plasma releasate injection in rabbit discs degenerated by intradiscal injection of condoliase. Arthritis Res. Ther., 2023, 25: 216.

[430]

Chiang ER. et al.. Use of allogeneic hypoxic mesenchymal stem cells for treating disc degeneration in rabbits. J. Orthop. Res., 2019, 37: 1440-1450.

[431]

Lu S, Lin CW. Lentivirus-mediated transfer of gene encoding fibroblast growth factor-18 inhibits intervertebral disc degeneration. Exp. Ther. Med., 2021, 22: 856.

[432]

Banala RR. et al.. Efficiency of dual siRNA-mediated gene therapy for intervertebral disc degeneration (IVDD). Spine J., 2019, 19: 896-904.

[433]

Mizrahi O. et al.. Nucleus pulposus degeneration alters properties of resident progenitor cells. Spine J., 2013, 13: 803-814.

[434]

Flouzat-Lachaniette CH. et al.. A novel in vivo porcine model of intervertebral disc degeneration induced by cryoinjury. Int. Orthop., 2018, 42: 2263-2272.

[435]

Yoon SH. et al.. A porcine model of intervertebral disc degeneration induced by annular injury characterized with magnetic resonance imaging and histopathological findings. Laboratory investigation. J. Neurosurg. Spine, 2008, 8: 450-457.

[436]

Kang R. et al.. Interference in the endplate nutritional pathway causes intervertebral disc degeneration in an immature porcine model. Int. Orthop., 2014, 38: 1011-1017.

[437]

Niinimaki J. et al.. Quantitative magnetic resonance imaging of experimentally injured porcine intervertebral disc. Acta Radio., 2007, 48: 643-649.

[438]

Omlor GW. et al.. A new porcine in vivo animal model of disc degeneration: response of anulus fibrosus cells, chondrocyte-like nucleus pulposus cells, and notochordal nucleus pulposus cells to partial nucleotomy. Spine (Philos. Pa 1976), 2009, 34: 2730-2739.

[439]

Acosta FLJr.. et al.. Porcine intervertebral disc repair using allogeneic juvenile articular chondrocytes or mesenchymal stem cells. Tissue Eng. Part A, 2011, 17: 3045-3055.

[440]

Lee, S. E. & Jahng, T. A. A new porcine animal model of cervical disc degeneration. Glob. Spine J.4, s-0034-1376582-s-1370034-1376582 (2014).

[441]

Henriksson HB. et al.. Transplantation of human mesenchymal stems cells into intervertebral discs in a xenogeneic porcine model. Spine (Philos. Pa 1976), 2009, 34: 141-148.

[442]

Omlor GW. et al.. Methods to monitor distribution and metabolic activity of mesenchymal stem cells following in vivo injection into nucleotomized porcine intervertebral discs. Eur. Spine J., 2010, 19: 601-612.

[443]

Cinotti G. et al.. Degenerative changes of porcine intervertebral disc induced by vertebral endplate injuries. Spine (Philos. Pa 1976), 2005, 30: 174-180.

[444]

Holm S. et al.. Pro-inflammatory, pleiotropic, and anti-inflammatory TNF-alpha, IL-6, and IL-10 in experimental porcine intervertebral disk degeneration. Vet. Pathol., 2009, 46: 1292-1300.

[445]

Detiger SE. et al.. MRI T2* mapping correlates with biochemistry and histology in intervertebral disc degeneration in a large animal model. Eur. Spine J., 2015, 24: 1935-1943.

[446]

Hoogendoorn RJ, Wuisman PI, Smit TH, Everts VE, Helder MN. Experimental intervertebral disc degeneration induced by chondroitinase ABC in the goat. Spine (Philos. Pa 1976), 2007, 32: 1816-1825.

[447]

Detiger SE. et al.. Biomechanical and rheological characterization of mild intervertebral disc degeneration in a large animal model. J. Orthop. Res., 2013, 31: 703-709.

[448]

Peeters M. et al.. BMP-2 and BMP-2/7 heterodimers conjugated to a fibrin/hyaluronic acid hydrogel in a large animal model of mild intervertebral disc degeneration. Biores Open Access, 2015, 4: 398-406.

[449]

Paul, C. P. L. et al. Changes in intervertebral disk mechanical behavior during early degeneration. J. Biomech. Eng.140. https://doi.org/10.1115/1.4039890 (2018).

[450]

Muir VG. et al.. Injectable radiopaque hyaluronic acid granular hydrogels for intervertebral disc repair. Adv. Health Mater., 2024, 13: e2303326.

[451]

Lim KZ. et al.. Ovine Lumbar intervertebral disc degeneration model utilizing a lateral retroperitoneal drill bit injury. J. Vis. Exp., 2017.

[452]

Pennicooke B. et al.. Annulus fibrosus repair using high-density collagen gel: an in vivo ovine model. Spine (Philos. Pa 1976), 2018, 43: E208-E215.

[453]

Ghosh P. et al.. Immunoselected STRO-3+ mesenchymal precursor cells and restoration of the extracellular matrix of degenerate intervertebral discs. J. Neurosurg. Spine, 2012, 16: 479-488.

[454]

Borem R. et al.. Characterization of chondroitinase-induced lumbar intervertebral disc degeneration in a sheep model intended for assessing biomaterials. J. Biomed. Mater. Res. A, 2021, 109: 1232-1246.

[455]

Gu T. et al.. Human bone morphogenetic protein 7 transfected nucleus pulposus cells delay the degeneration of intervertebral disc in dogs. J. Orthop. Res., 2017, 35: 1311-1322.

[456]

Matta A. et al.. NTG-101: a novel molecular therapy that halts the progression of degenerative disc disease. Sci. Rep., 2018, 8. 16809

[457]

Zeng Y. et al.. Injectable microcryogels reinforced alginate encapsulation of mesenchymal stromal cells for leak-proof delivery and alleviation of canine disc degeneration. Biomaterials, 2015, 59: 53-65.

[458]

Li W. et al.. Blocking the function of inflammatory cytokines and mediators by using IL-10 and TGF-beta: a potential biological immunotherapy for intervertebral disc degeneration in a beagle model. Int. J. Mol. Sci., 2014, 15: 17270-17283.

[459]

Shi Z. et al.. Intervention of rAAV-hTERT-transducted nucleus pulposus cells in early stage of intervertebral disc degeneration: a study in canine model. Tissue Eng. Part A, 2015, 21: 2186-2194.

[460]

Hutton WC. et al.. Does long-term compressive loading on the intervertebral disc cause degeneration?. Spine (Philos. Pa 1976), 2000, 25: 2993-3004.

[461]

Hutton WC. et al.. The effect of compressive force applied to the intervertebral disc in vivo. A study of proteoglycans and collagen. Spine (Philos. Pa 1976), 1998, 23: 2524-2537.

[462]

Hiyama A. et al.. Transplantation of mesenchymal stem cells in a canine disc degeneration model. J. Orthop. Res., 2008, 26: 589-600.

[463]

Pfeiffer M. et al.. Intradiscal application of hyaluronic acid in the non-human primate lumbar spine: radiological results. Eur. Spine J., 2003, 12: 76-83.

[464]

Wei F. et al.. In vivo experimental intervertebral disc degeneration induced by bleomycin in the rhesus monkey. BMC Musculoskelet. Disord., 2014, 15. 340

[465]

Stern WE, Coulson WF. Effects of collagenase upon the intervertebral disc in monkeys. J. Neurosurg., 1976, 44: 32-44.

[466]

Zook BC, Kobrine AI. Effects of collagenase and chymopapain on spinal nerves and intervertebral discs of cynomolgus monkeys. J. Neurosurg., 1986, 64: 474-483.

[467]

Chen JX. et al.. Annular defects impair the mechanical stability of the intervertebral disc. Glob. Spine J., 2023, 13: 724-729.

[468]

Russo, F. et al. A hyaluronan and platelet-rich plasma hydrogel for mesenchymal stem cell delivery in the intervertebral disc: an organ culture study. Int. J. Mol. Sci.22 (2021). https://doi.org/10.3390/ijms220629634

[469]

de Souza Grava AL, Ferrari LF, Parada CA, Defino HL. Pharmacologic treatment of hyperalgesia experimentally induced by nucleus pulposus. Rev. Bras. Ortop., 2010, 45: 569-576

[470]

Lee S. et al.. Voluntary running attenuates behavioural signs of low back pain: dimorphic regulation of intervertebral disc inflammation in male and female SPARC-null mice. Osteoarthr. Cartil., 2022, 30: 110-123.

[471]

Lillyman DJ. et al.. Axial hypersensitivity is associated with aberrant nerve sprouting in a novel model of disc degeneration in female Sprague Dawley rats. JOR Spine, 2022, 5. e1212

[472]

Lee FS, Cruz CJ, Allen KD, Wachs RA. Gait assessment in a female rat Sprague Dawley model of disc-associated low back pain. Connect Tissue Res., 2024, 65: 407-420.

[473]

Pelled G. et al.. Intradiscal quantitative chemical exchange saturation transfer MRI signal correlates with discogenic pain in human patients. Sci. Rep., 2021, 11. 19195

[474]

Schleich C. et al.. Glycosaminoglycan chemical exchange saturation transfer of lumbar intervertebral discs in healthy volunteers. Spine (Philos. Pa 1976), 2016, 41: 146-152.

[475]

Haneder S. et al.. Assessment of glycosaminoglycan content in intervertebral discs using chemical exchange saturation transfer at 3.0 Tesla: preliminary results in patients with low-back pain. Eur. Radio., 2013, 23: 861-868.

[476]

Aliyev A. et al.. Age-related inflammatory changes in the spine as demonstrated by (18)F-FDG-PET:observation and insight into degenerative spinal changes. Hell. J. Nucl. Med., 2012, 15: 197-201

[477]

Zhou X. et al.. Detection of nociceptive-related metabolic activity in the spinal cord of low back pain patients using (18)F-FDG PET/CT. Scand. J. Pain., 2017, 15: 53-57.

[478]

Gamie S, El-Maghraby T. The role of PET/CT in evaluation of facet and disc abnormalities in patients with low back pain using (18)F-Fluoride. Nucl. Med. Rev. Cent. East Eur., 2008, 11: 17-21

[479]

Sharma DN, Yerramneni VK, Srivastava MK, Yerragunta T, Akurati S. Role of magnetic resonance imaging and 18-fluorodeoxyglucose positron emission tomography-computed tomography in identifying pain generators in patients with chronic low back pain. J. Craniovertebr Junction Spine, 2023, 14: 381-387.

[480]

Piri R. et al.. PET/CT imaging of spinal inflammation and microcalcification in patients with low back pain: a pilot study on the quantification by artificial intelligence-based segmentation. Clin. Physiol. Funct. Imaging, 2022, 42: 225-232.

[481]

Gornet MG. et al.. Magnetic resonance spectroscopy (MRS) can identify painful lumbar discs and may facilitate improved clinical outcomes of lumbar surgeries for discogenic pain. Eur. Spine J., 2019, 28: 674-687.

[482]

Wilson L, Beall DP, Eastlack RK, Berven S, Lotz JC. The comparison of cost-effectiveness between magnetic resonance spectroscopy and provocative discography in the identification of chronic low back pain surgery candidates. Clinicoecon Outcomes Res., 2025, 17: 19-31.

Funding

U.S. Department of Health & Human Services | NIH | National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)(R01AR082041)

California Institute for Regenerative Medicine (CIRM)(DISC2-14049)

RIGHTS & PERMISSIONS

The Author(s)

AI Summary AI Mindmap
PDF

111

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/