Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Elevated dietary ω-6 polyunsaturated fatty acids induce reversible peripheral nerve dysfunction that exacerbates comorbid pain conditions

Abstract

Chronic pain is the leading cause of disability worldwide1 and is commonly associated with comorbid disorders2. However, the role of diet in chronic pain is poorly understood. Of particular interest is the Western-style diet, enriched with ω-6 polyunsaturated fatty acids (PUFAs) that accumulate in membrane phospholipids and oxidise into pronociceptive oxylipins3,4. Here we report that mice administered an ω-6 PUFA-enriched diet develop persistent nociceptive hypersensitivities, spontaneously active and hyper-responsive glabrous afferent fibres and histologic markers of peripheral nerve damage reminiscent of a peripheral neuropathy. Linoleic and arachidonic acids accumulate in lumbar dorsal root ganglia, with increased liberation via elevated phospholipase (PLA)2 activity. Pharmacological and molecular inhibition of PLA2G7 or diet reversal with high levels of ω-3 PUFAs attenuate nociceptive behaviours, neurophysiologic abnormalities and afferent histopathology induced by high ω-6 intake. Additionally, ω-6 PUFA accumulation exacerbates allodynia observed in preclinical inflammatory and neuropathic pain models and is strongly correlated with multiple pain indices of clinical diabetic neuropathy. Collectively, these data reveal dietary enrichment with ω-6 PUFAs as a new aetiology of peripheral neuropathy and risk factor for chronic pain and implicate multiple therapeutic considerations for clinical pain management.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: An ω-6 fatty acid–enriched diet induces a peripheral neuropathy–like phenotype in mice.
Fig. 2: The H6D increases membrane loading of ω-6 PUFAs and stimulates PLA2 activity in peripheral afferent neurons.
Fig. 3: An ω-3 fatty acid–enriched diet rescues the H6D-induced neuropathy-like phenotype.
Fig. 4: Diet-specific modulation of nociceptive behaviours associated with inflammatory and neuropathic pain.

Similar content being viewed by others

Data availability

Data that support the findings of this study are available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Vos, T. et al. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet 390, 1211–1259 (2017).

    Article  Google Scholar 

  2. Mills, S. E. E., Nicolson, K. P. & Smith, B. H. Chronic pain: a review of its epidemiology and associated factors in population-based studies. Br. J. Anaesth. 123, e273–e283 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Gabbs, M., Leng, S., Devassy, J. G., Monirujjaman, M. & Aukema, H. M. Advances in our understanding of oxylipins derived from dietary PUFAs. Adv. Nutr. 6, 513–540 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Shearer, G. C. & Walker, R. E. An overview of the biologic effects of omega-6 oxylipins in humans. Prostaglandins Leukot. Essent. Fatty Acids 137, 26–38 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Simopoulos, A. P. Evolutionary aspects of diet, the omega-6/omega-3 ratio and genetic variation: nutritional implications for chronic diseases. Biomed. Pharmacother. 60, 502–507 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Mann, J. I. Diet and risk of coronary heart disease and type 2 diabetes. Lancet 360, 783–789 (2002).

    Article  CAS  PubMed  Google Scholar 

  7. Manzel, A. et al. Role of ‘western diet’ in inflammatory autoimmune diseases. Curr. Allergy Asthma Rep. 14, 404 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Blasbalg, T. L., Hibbeln, J. R., Ramsden, C. E., Majchrzak, S. F. & Rawlings, R. R. Changes in consumption of omega-3 and omega-6 fatty acids in the United States during the 20th century. Am. J. Clin. Nutr. 93, 950–962 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kris-Etherton, P. M. et al. Polyunsaturated fatty acids in the food chain in the United States. Am. J. Clin. Nutr. 71, 179S–188S (2000).

    Article  CAS  PubMed  Google Scholar 

  10. Patwardhan, A. M. et al. Heat generates oxidized linoleic acid metabolites that activate TRPV1 and produce pain in rodents. J. Clin. Invest. 120, 1617–1626 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Eskander, M. A. et al. Persistent nociception triggered by nerve growth factor (NGF) is mediated by TRPV1 and oxidative mechanisms. J. Neurosci. 35, 8593–8603 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Green, D. P., Ruparel, S., Roman, L., Henry, M. A. & Hargreaves, K. M. Role of endogenous TRPV1 agonists in a postburn pain model of partial-thickness injury. Pain 154, 2512–2520 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Ramsden, C. E. et al. A systems approach for discovering linoleic acid derivatives that potentially mediate pain and itch. Sci. Signal. 10, eaal5241 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Sisignano, M. et al. Targeting CYP2J to reduce paclitaxel-induced peripheral neuropathic pain. Proc. Natl Acad. Sci. USA 113, 12544–12549 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Clarke, G. et al. Marked elevations in pro-inflammatory polyunsaturated fatty acid metabolites in females with irritable bowel syndrome. J. Lipid Res. 51, 1186–1192 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Adam, O. et al. Anti-inflammatory effects of a low arachidonic acid diet and fish oil in patients with rheumatoid arthritis. Rheumatol. Int. 23, 27–36 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Calder, P. C. Session 3: Joint Nutrition Society and Irish Nutrition and Dietetic Institute Symposium on ‘Nutrition and autoimmune disease’ PUFA, inflammatory processes and rheumatoid arthritis. Proc. Nutr. Soc. 67, 409–418 (2008).

    Article  CAS  PubMed  Google Scholar 

  18. Devigili, G. et al. The diagnostic criteria for small fibre neuropathy: from symptoms to neuropathology. Brain 131, 1912–1925 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Lauria, G. et al. European Federation of Neurological Societies/Peripheral Nerve Society Guideline on the use of skin biopsy in the diagnosis of small fiber neuropathy. Report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society. Eur. J. Neurol. 17, 903–912 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Hunt, D., Raivich, G. & Anderson, P. N. Activating transcription factor 3 and the nervous system. Front. Mol. Neurosci. 5, 7 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tsujino, H. et al. Activating transcription factor 3 (ATF3) induction by axotomy in sensory and motoneurons: a novel neuronal marker of nerve injury. Mol. Cell. Neurosci. 15, 170–182 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Bray, G. A. & Popkin, B. M. Dietary fat intake does affect obesity! Am. J. Clin. Nutr. 68, 1157–1173 (1998).

    Article  CAS  PubMed  Google Scholar 

  23. Forouhi, N. G. et al. Association of plasma phospholipid n-3 and n-6 polyunsaturated fatty acids with type 2 diabetes: the EPIC-InterAct case-cohort study. PLoS Med. 13, e1002094 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Fung, T. T., Schulze, M., Manson, J. E., Willett, W. C. & Hu, F. B. Dietary patterns, meat intake, and the risk of type 2 diabetes in women. Arch. Intern. Med. 164, 2235–2240 (2004).

    Article  PubMed  Google Scholar 

  25. Mente, A. et al. Association of dietary nutrients with blood lipids and blood pressure in 18 countries: a cross-sectional analysis from the PURE study. Lancet Diabetes Endocrinol. 5, 774–787 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. van Dam, R. M., Willett, W. C., Rimm, E. B., Stampfer, M. J. & Hu, F. B. Dietary fat and meat intake in relation to risk of type 2 diabetes in men. Diabetes Care 25, 417–424 (2002).

    Article  PubMed  Google Scholar 

  27. Ramsden, C. E. et al. Dietary linoleic acid-induced alterations in pro- and anti-nociceptive lipid autacoids: implications for idiopathic pain syndromes? Mol. Pain 12, 1744806916636386 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Patwardhan, A. M., Scotland, P. E., Akopian, A. N. & Hargreaves, K. M. Activation of TRPV1 in the spinal cord by oxidized linoleic acid metabolites contributes to inflammatory hyperalgesia. Proc. Natl Acad. Sci. USA 106, 18820–18824 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Griffin, T. M. et al. Diet-induced obesity differentially regulates behavioral, biomechanical, and molecular risk factors for osteoarthritis in mice. Arthritis Res. Ther. 12, R130 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Lee, E. et al. Transient receptor potential vanilloid type-1 channel regulates diet-induced obesity, insulin resistance, and leptin resistance. FASEB J. 29, 3182–3192 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tramullas, M., Finger, B. C., Dinan, T. G. & Cryan, J. F. Obesity takes its toll on visceral pain: high-fat diet induces Toll-like receptor 4-dependent visceral hypersensitivity. PLoS ONE 11, e0155367 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Wilensky, R. L. et al. Inhibition of lipoprotein-associated phospholipase A2 reduces complex coronary atherosclerotic plaque development. Nat. Med. 14, 1059–1066 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wallace, V. C., Cottrell, D. F., Brophy, P. J. & Fleetwood-Walker, S. M. Focal lysolecithin-induced demyelination of peripheral afferents results in neuropathic pain behavior that is attenuated by cannabinoids. J. Neurosci. 23, 3221–3233 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Piomelli, D. & Sasso, O. Peripheral gating of pain signals by endogenous lipid mediators. Nat. Neurosci. 17, 164–174 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Dennis, E. A., Cao, J., Hsu, Y. H., Magrioti, V. & Kokotos, G. Phospholipase A2 enzymes: physical structure, biological function, disease implication, chemical inhibition, and therapeutic intervention. Chem. Rev. 111, 6130–6185 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Usoskin, D. et al. Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencing. Nat. Neurosci. 18, 145–153 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Kim, M. et al. Impact of 8-week linoleic acid intake in soy oil on Lp-PLA2 activity in healthy adults. Nutr. Metab. 14, 32 (2017).

    Article  Google Scholar 

  38. Blackie, J. A. et al. The identification of clinical candidate SB-480848: a potent inhibitor of lipoprotein-associated phospholipase A2. Bioorg. Med. Chem. Lett. 13, 1067–1070 (2003).

    Article  CAS  PubMed  Google Scholar 

  39. Goldberg, R. J. & Katz, J. A meta-analysis of the analgesic effects of omega-3 polyunsaturated fatty acid supplementation for inflammatory joint pain. Pain 129, 210–223 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Laye, S., Nadjar, A., Joffre, C. & Bazinet, R. P. Anti-inflammatory effects of omega-3 fatty acids in the brain: physiological mechanisms and relevance to pharmacology. Pharmacol. Rev. 70, 12–38 (2018).

    Article  CAS  PubMed  Google Scholar 

  41. Xu, Z. Z. et al. Resolvins RvE1 and RvD1 attenuate inflammatory pain via central and peripheral actions. Nat. Med. 16, 592–597 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bazata, D. D., Robinson, J. G., Fox, K. M., Grandy, S. & Group, S. S. Affecting behavior change in individuals with diabetes: findings from the Study to Help Improve Early Evaluation and Management of Risk Factors Leading to Diabetes (SHIELD). Diabetes Educ. 34, 1025–1036 (2008).

    Article  PubMed  Google Scholar 

  43. Davis, J. A., Robinson, R. L., Le, T. K. & Xie, J. Incidence and impact of pain conditions and comorbid illnesses. J. Pain Res. 4, 331–345 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Fehrenbacher, J. C., Vasko, M. R. & Duarte, D. B. Models of inflammation: carrageenan- or complete Freund’s adjuvant (CFA)-induced edema and hypersensitivity in the rat. Curr. Protoc. Pharmacol. 56, 5.4 (2012).

    Article  Google Scholar 

  45. Kobayashi, K. et al. The db/db mouse, a model for diabetic dyslipidemia: molecular characterization and effects of Western diet feeding. Metabolism 49, 22–31 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. O’Brien, P. D., Sakowski, S. A. & Feldman, E. L. Mouse models of diabetic neuropathy. ILAR J. 54, 259–272 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Alvheim, A. R. et al. Dietary linoleic acid elevates endogenous 2-AG and anandamide and induces obesity. Obesity 20, 1984–1994 (2012).

    Article  CAS  PubMed  Google Scholar 

  48. Bennett, M. The LANSS Pain Scale: the Leeds assessment of neuropathic symptoms and signs. Pain 92, 147–157 (2001).

    Article  CAS  PubMed  Google Scholar 

  49. Bouhassira, D. et al. Development and validation of the Neuropathic Pain Symptom Inventory. Pain 108, 248–257 (2004).

    Article  PubMed  Google Scholar 

  50. Mauck, M. C. et al. Obesity increases the risk of chronic pain development after motor vehicle collision. Pain 160, 670–675 (2019).

    Article  PubMed  Google Scholar 

  51. Okifuji, A. & Hare, B. D. The association between chronic pain and obesity. J. Pain Res. 8, 399–408 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  52. White, H. D. et al. Darapladib for preventing ischemic events in stable coronary heart disease. N. Engl. J. Med. 370, 1702–1711 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. O’Donoghue, M. L. et al. Effect of darapladib on major coronary events after an acute coronary syndrome: the SOLID-TIMI 52 randomized clinical trial. JAMA 312, 1006–1015 (2014).

    Article  PubMed  Google Scholar 

  54. Haghdoost, F. et al. Association between Ala379Val polymorphism of lipoprotein-associated phospholipase A2 and migraine without aura in Iranian population. Iran. J. Neurol. 15, 80–84 (2016).

    PubMed  PubMed Central  Google Scholar 

  55. Parisien, M. et al. Effect of human genetic variability on gene expression in dorsal root ganglia and association with pain phenotypes. Cell Rep. 19, 1940–1952 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Hummel, K. P., Dickie, M. M. & Coleman, D. L. Diabetes, a new mutation in the mouse. Science 153, 1127–1128 (1966).

    Article  CAS  PubMed  Google Scholar 

  57. Reeves, P. G., Nielsen, F. H. & Fahey, G. C. Jr. AIN-93 purified diets for laboratory rodents: final report of the American Institute of Nutrition ad hoc writing committee on the reformulation of the AIN-76A rodent diet. J. Nutr. 123, 1939–1951 (1993).

    Article  CAS  PubMed  Google Scholar 

  58. Gibbs, J. L., Flores, C. M. & Hargreaves, K. M. Attenuation of capsaicin-evoked mechanical allodynia by peripheral neuropeptide Y Y1 receptors. Pain 124, 167–174 (2006).

    Article  CAS  PubMed  Google Scholar 

  59. Chaplan, S. R., Bach, F. W., Pogrel, J. W., Chung, J. M. & Yaksh, T. L. Quantitative assessment of tactile allodynia in the rat paw. J. Neurosci. Methods 53, 55–63 (1994).

    Article  CAS  PubMed  Google Scholar 

  60. White, S., Marquez de Prado, B., Russo, A. F. & Hammond, D. L. Heat hyperalgesia and mechanical hypersensitivity induced by calcitonin gene-related peptide in a mouse model of neurofibromatosis. PLoS ONE 9, e106767 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Garrison, S. R., Dietrich, A. & Stucky, C. L. TRPC1 contributes to light-touch sensation and mechanical responses in low-threshold cutaneous sensory neurons. J. Neurophysiol. 107, 913–922 (2012).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  63. Brenner, D. S., Golden, J. P. & Gereau, R. W. T. A novel behavioral assay for measuring cold sensation in mice. PLoS ONE 7, e39765 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ayala, J. E. et al. Standard operating procedures for describing and performing metabolic tests of glucose homeostasis in mice. Dis. Model. Mech. 3, 525–534 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Han, B. G. et al. Markers of glycemic control in the mouse: comparisons of 6-h- and overnight-fasted blood glucoses to Hb A1c. Am. J. Physiol. Endocrinol. Metab. 295, E981–E986 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Wang, M. & Han, X. Multidimensional mass spectrometry-based shotgun lipidomics. Methods Mol. Biol. 1198, 203–220 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Han, X., Yang, K. & Gross, R. W. Microfluidics-based electrospray ionization enhances the intrasource separation of lipid classes and extends identification of individual molecular species through multi-dimensional mass spectrometry: development of an automated high-throughput platform for shotgun lipidomics. Rapid Commun. Mass Spectrom. 22, 2115–2124 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Wang, M., Wang, C., Han, R. H. & Han, X. Novel advances in shotgun lipidomics for biology and medicine. Prog. Lipid Res. 61, 83–108 (2016).

    Article  CAS  PubMed  Google Scholar 

  69. Yang, K., Cheng, H., Gross, R. W. & Han, X. Automated lipid identification and quantification by multidimensional mass spectrometry-based shotgun lipidomics. Anal. Chem. 81, 4356–4368 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Bligh, E. G. & Dyer, W. J. A rapid method of total lipid extraction and purification. Can. J. Biochem. Physiol. 37, 911–917 (1959).

    Article  CAS  PubMed  Google Scholar 

  71. Pettinella, C., Lee, S. H., Cipollone, F. & Blair, I. A. Targeted quantitative analysis of fatty acids in atherosclerotic plaques by high sensitivity liquid chromatography/tandem mass spectrometry. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 850, 168–176 (2007).

    Article  CAS  PubMed  Google Scholar 

  72. Quehenberger, O., Armando, A. M. & Dennis, E. A. High sensitivity quantitative lipidomics analysis of fatty acids in biological samples by gas chromatography–mass spectrometry. Biochim. Biophys. Acta 1811, 648–656 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Reeh, P. W. Sensory receptors in a mammalian skin-nerve in vitro preparation. Prog. Brain Res. 74, 271–276 (1988).

    Article  CAS  PubMed  Google Scholar 

  74. Banik, R. K. & Brennan, T. J. Sensitization of primary afferents to mechanical and heat stimuli after incision in a novel in vitro mouse glabrous skin-nerve preparation. Pain 138, 380–391 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Zimmermann, K. et al. Phenotyping sensory nerve endings in vitro in the mouse. Nat. Protoc. 4, 174–196 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Hogan, D., Baker, A. L., Moron, J. A. & Carlton, S. M. Systemic morphine treatment induces changes in firing patterns and responses of nociceptive afferent fibers in mouse glabrous skin. Pain 154, 2297–2309 (2013).

    Article  CAS  PubMed  Google Scholar 

  77. Moehring, F. et al. Keratinocytes mediate innocuous and noxious touch via ATP–P2X4 signaling. eLife 7, e31684 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Duraku, L. S. et al. Spatiotemporal dynamics of re-innervation and hyperinnervation patterns by uninjured CGRP fibers in the rat foot sole epidermis after nerve injury. Mol. Pain 8, 61 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Reid, G., Amuzescu, B., Zech, E. & Flonta, M. L. A system for applying rapid warming or cooling stimuli to cells during patch clamp recording or ion imaging. J. Neurosci. Methods 111, 1–8 (2001).

    Article  CAS  PubMed  Google Scholar 

  80. Koltzenburg, M., Stucky, C. L. & Lewin, G. R. Receptive properties of mouse sensory neurons innervating hairy skin. J. Neurophysiol. 78, 1841–1850 (1997).

    Article  CAS  PubMed  Google Scholar 

  81. Banik, R. K. & Brennan, T. J. Spontaneous discharge and increased heat sensitivity of rat C-fiber nociceptors are present in vitro after plantar incision. Pain 112, 204–213 (2004).

    Article  PubMed  Google Scholar 

  82. Collins, T. J. ImageJ for microscopy. Biotechniques 43, 25–30 (2007).

    Article  PubMed  Google Scholar 

  83. Linkert, M. et al. Metadata matters: access to image data in the real world. J. Cell Biol. 189, 777–782 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  85. Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 9, 671–675 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. LoCoco, P. M. et al. Pharmacological augmentation of nicotinamide phosphoribosyltransferase (NAMPT) protects against paclitaxel-induced peripheral neuropathy. eLife 6, e29626 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  87. Beiswenger, K. K., Calcutt, N. A. & Mizisin, A. P. Epidermal nerve fiber quantification in the assessment of diabetic neuropathy. Acta Histochem. 110, 351–362 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  88. LoCoco, P. M. et al. Reliable approaches to extract high-integrity RNA from skin and other pertinent tissues used in pain research. Pain Rep. 5, e818 (2020).

  89. Livak, K. J. & Schmittgen, T. D. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔC(T) method. Methods 25, 402–408 (2001).

    Article  CAS  PubMed  Google Scholar 

  90. Harris, P. A. et al. Research electronic data capture (REDCap)—a metadata-driven methodology and workflow process for providing translational research informatics support. J. Biomed. Inform. 42, 377–381 (2009).

    Article  PubMed  Google Scholar 

  91. Beery, A. K. Inclusion of females does not increase variability in rodent research studies. Curr. Opin. Behav. Sci. 23, 143–149 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The project described was supported in part by the National Center for Advancing Translational Sciences, National Institutes of Health (NIH), through grant UL1TR002645 (K.M.H.). Additional support from the NIH includes grants R01NS110948 (K.M.H.), T32DE14318 (P.M.L., A.R.F., K.M.H.), T32GM113896 (J.T.B.), F30AT009949 (J.T.B.), F32DK118841 (P.M.L.), F30DE028486 (A.R.F.) and a grant from the Ella and Williams Owen’s Foundation (K.M.H.). Clinical data were managed using REDCap software supported by UL1RR024982. Certain mass spectrometric analyses were carried out on equipment supported by the US Department of Agriculture, Agricultural Research Service, under agreement no. 58-3094-8-012. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH or the US Department of Agriculture. We thank X. Han and his laboratory for expertise and guidance on shotgun lipidomics. We thank M. Patil and P. Wu for technical assistance as well as A. Diogenes, N. Ruparel, A. Khan and A. Akopian for fruitful discussions.

Author information

Authors and Affiliations

Authors

Contributions

J.T.B., P.M.L., S.B.R. and K.M.H. conceived and designed the studies; J.T.B., P.M.L., M.R.B. and M.T. conducted behavioural experiments; A.R.F., P.M.L. and F.-M.C. conducted single-fibre electrophysiology; P.M.L. and J.T.B. performed histology; Q.L., F.-M.C. and P.M.L. performed BODIPY experiments; P.M.L., D.A.A. and M.T. performed total tissue lipid extractions; M.E.C., G.M.S. and S.B.H.B. conducted quantitative LC–MS/MS; P.M.L. and F.-M.C. performed western blots; E.E.L. and A.T. conducted neurological assessments on trial participants and collected skin punch biopsies; J.T.B., P.M.L. and K.M.H. performed data analysis for all experiments; P.M.L. and J.T.B. prepared figures, images and illustrations; P.M.L., J.T.B. and K.M.H. wrote the manuscript; all authors revised the manuscript.

Corresponding author

Correspondence to Kenneth M. Hargreaves.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Jing Kang, Ru-Rong Ji and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Christoph Schmitt.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 H6D induces persistent nociceptive hypersensitivities in both male and female mice.

a-f, Time courses of changes in mechanical withdrawal threshold (a,c,e) and heat withdrawal latency (b,d,f) for male and female mice on the H6D and L6D. The top two plots (a,b) represent compiled male and female responses (L6D, n = 24; H6D, n = 29). a, ***P = 0.0003 week 4, ****P < 0.0001 weeks 8–24. b, ****P < 0.0001 weeks 4–24. The middle plots (c,d) are male-only responses (L6D, n = 12; H6D, n = 16). c, *P = 0.0401 week 4, ****P < 0.0001 weeks 8–24. d, ****P < 0.0001 weeks 4–24. The bottom plots (e,f) are female-only responses (L6D, n = 12; H6D, n = 13). e, **P = 0.0055 week 16, 0.0023 week 20, ***P = 0.0009 week 4, 0.0005 week 8, ****P < 0.0001 weeks 12,24. f, **P = 0.0022 week 4, ****P < 0.0001 weeks 8–24. Data are mean ± SEM. Error bars for some data points are within the size of the symbol. The statistical test used was two-way repeated-measures ANOVA with Sidak’s post-hoc test (a-f).

Source data

Extended Data Fig. 2 The H6D sensitizes afferent fibers to mechanical and heat stimuli.

a, Representative recording wavemarks from L6D and H6D mice during mechanical force application. The number of action potentials are denoted beneath each stimulation for each recording. b, Percentage of fibers exhibiting post-stimulus afterdischarge following mechanical force application. Values represent the number of fibers exhibiting afterdischarge over the total recorded fibers for each group (L6D, n = 34; H6D, n = 44). **P = 0.0096. c, The Peltier-based heat delivery system setup. d, Conduction velocities determined for recorded C (left) and AM (right) fibers from L6D and H6D mice (L6D-C, n = 19; H6D-C, n = 20; L6D-AM, n = 25; H6D-AM, n = 24). Dotted line represents cut-off value for C fiber classification. e,f, Glabrous IENF densities (e) and percentage of ATF3+ neurons in lumbar DRG (f) after 4 weeks on the L6D or H6D (L6D, n = 3; H6D, n = 3). g, Representative immunofluorescent staining of ATF3 expression in trigeminal ganglia (TG) from L6D and H6D mice co-localized with NeuN, scale bars: 50 μm (n = 2 mice/group). h,i, Immunofluorescent staining of Iba1 (h) and c-Fos (i) expression in the lumbar spinal cord of L6D and H6D mice. Positive control tissue was utilized from db/db mice. White arrowheads designate microglia (h), 10X magnification, scale bar: 50 μm (n = 2 mice/group). Red arrowheads designate c-Fos+ nuclei (i), 20X magnification, scale bar: 50 μm (n = 2 mice/group). All data are mean ± SEM. The statistical test used was a two-sided Fisher’s exact test (b).

Source data

Extended Data Fig. 3 The H6D does not induce a diabetic phenotype.

a,b, Scatter plots of fasting blood glucose levels (a) and HbA1c levels (b) from mice on L6D and H6D for 8 weeks. Mice on normal chow (NC) and 16-week-old db/db mice served as negative and positive controls, respectively (NC, n = 5; L6D, n = 5; H6D, n = 5; db/db, n = 5). Dotted lines in each figure represent established cut-offs for type 2 diabetes. ****P < 0.0001 (db/db vs NC). c,d, Weekly monitoring of body weights (c) and food intake (d) for both male and female mice on either L6D or H6D. Data are mean ± SEM. Error bars for some data points are within the size of the symbol. Statistical test used was one-way ANOVA with Tukey’s post-hoc test (a,b).

Source data

Extended Data Fig. 4 The H6D alters lipid composition in DRG, but not spinal cord.

a,b, Heatmaps of lipid species from lumbar DRG (a) and spinal cord (b) from male (♂) and female (♀) mice on either the H6D or L6D. Lipid classes are designated to the left of each heatmap. Scale bar represents z-score transformations for each lipid species. c,d, Scatter plots of LA- and AA-esterified lipids (c) as well as ω-3 content (d, EPA, DHA levels) in DRG sub-profiled by lipid class for male and female mice on either diet (n = 3 mice/group/sex). c, Acyl carnitine (LA): *P = 0.0346, ***P = 0.0004. Acyl carnitine (AA): *P = 0.0417 (♂), *P = 0.0214 (♀). Ethanolamine plasmalogens (LA): ***P = 0.0008, ****P < 0.0001. Ethanolamine plasmalogens (AA): **P = 0.0027, ***P = 0.0007. Fatty acyl chains (LA): **P = 0.0069. Lyso-phosphatidylcholine (LA): ***P = 0.0002, ****P < 0.0001. Lyso-phosphatidylcholine (AA): *P = 0.0161, *P = 0.0215. Lyso-phosphatidylethanolamine (LA): ****P < 0.0001. Phosphatidic acid (LA): **P = 0.005, *P = 0.0166. Phosphatidic acid (AA): ****P < 0.0001, ***P = 0.0001. Phosphatidylethanolamine (LA): ***P = 0.0006, ***P = 0.0002. Phosphatidylethanolamine (AA): **P = 0.0015, ***P = 0.0007. Phosphatidylglycerol (AA): **P = 0.0033. Phosphatidylinositol (AA): **P = 0.003, ***P = 0.001. Phosphatidylserine (LA): ***P = 0.0001, **P = 0.0072. d, Lyso-phosphatidylcholine: *P = 0.0316. Phosphatidylglycerol: **P = 0.0062. Data are mean ± SEM. Statistical tests used were two-way ANOVA with Tukey’s post-hoc test (c) and one-way ANOVA with Tukey’s post-hoc test (d).

Source data

Extended Data Fig. 5 PLA2g7 expression predominates in neuronal subpopulations of the lumbar DRG.

a, Heatmap indicating PLA2 isozyme expression across established neuronal subpopulations of the mouse lumbar DRG. Single-cell RNA-seq data were reproduced with permission36. b,c, qPCR data showing PLA2 isozyme expression (b) and change in cycle threshold values relative to 18 S rRNA (c) in lumbar DRG from H6D and L6D mice (b: n = 3/group. c: L6D, n = 6; H6D, n = 6). d, Representative immunofluorescent staining of PLA2g7 expression in mouse lumbar DRG and co-localization with neuronal subtype-specific markers. No primary controls are included for each marker as designated. The magenta arrows highlight two small diameter neurons, one with high PLA2g7 expression and one with low to moderate expression, that are negative for NFH. White arrows designate cell bodies with PLA2g7+ staining and their co-localization with the respective subtype marker. White arrowheads highlight axons projecting through the ganglia that exhibit virtually no PLA2g7 expression compared to cell bodies. Scale bars: 50 μm (n = 2 mice). e, Representative immunofluorescent staining of lumbar DRG from naïve control mice that received either scrambled or PLA2g7-directed siRNA intrathecally for the purpose of PLA2g7 antibody validation. White arrows highlight PLA2g7 + staining of neuronal cell bodies, whereas the white arrowheads designate cells exhibiting loss of PLA2g7 immunoreactivity. Scale bars: 50 μm, (n = 2 mice/group). f,g, Circulating plasma PLA2g7 levels (f) and plasma LA accumulation (g) from L6D and H6D mice after 8 weeks (L6D, n = 4; H6D, n = 5). **P < 0.0022. Data are mean ± SEM. Error bars for some data points are within the size of the symbol. Statistical test used was unpaired two-tailed Student’s t test (g).

Source data

Extended Data Fig. 6 Pharmacological inhibition and genetic knockdown of PLA2g7 in DRG neurons reduces PLA2 activity and attenuates nociceptive hypersensitivities.

a, Optimization of DRG protein concentration used with the PLA2 BODIPY activity assay. H6D DRG homogenates demonstrate increased activity at multiple concentrations compared to L6D (BSA, n = 4; L6D-300, n = 6; L6D-150, n = 7; L6D-75, n = 7; H6D-300, n = 5; H6D-150, n = 7; H6D-75, n = 7 DRG replicates/group). **P = 0.0055 (300), 0.0023 (150). b, qPCR data showing annexin isozyme expression in L6D and H6D DRG (L6D, n = 6; H6D, n = 5). c, Immunoblots of PLA2g7 and GAPDH protein expression in membrane and cytosolic fractions from homogenized DRG (L6D, n = 4 mice; H6D, n = 3 mice). Molecular weight markers (kDa) are adjacent to each target. d, Concentration-response curves for darapladib-mediated inhibition of PLA2 activity for DRG (n = 3/group). e, Darapladib half-maximal inhibitory concentrations (IC50) as determined by nonlinear regression (n = 3/group). f, Total LA and AA levels determined from glabrous hindpaw skin punches (L6D, n = 6; H6D, n = 6). **P = 0.0098 (LA), 0.0071 (AA). g, Dose-response timecourses for i.pl. darapladib on heat- and mechanical-evoked nociception (L6D-VEH, n = 6; L6D-3, n = 4; L6D-30, n = 9; H6D-VEH, n = 6; H6D-3, n = 5; H6D-30, n = 9). h,i, PLA2g7 qPCR data for lumbar DRG (h) and spinal cord (i) following intrathecal siRNA treatment (q.d. x 3d) (h: L6D-scr, n = 7; H6D-scr, n = 6; L6D-PLA2g7, n = 8; H6D-PLA2g7, n = 6. i: n = 3/group). h, *P = 0.0429 (L6D-PLA2g7), 0.0371 (H6D-PLA2g7). i, *P = 0.0333. j, Immunofluorescent images of glabrous IENFs from siRNA-treated mice, scale bar: 50 μm (n = 2 mice/group). k,l, Mechanical force-response curves (k) and EF50 values (l) for 16-week db/db mice injected i.pl. with either vehicle or darapladib (db/db-veh, n = 5; db/db-DARA, n = 6). l, **P = 0.0020. m,n, Mechanical force-response curves (m) and EF50 values (n) for a different cohort of 16-week db/db mice following i.t. siRNA injections (db/db-veh, n = 5; db/db-darapladib, n = 5). n, **P = 0.0066. All data are mean ± SEM. Error bars for some data points are within the size of the symbol. Statistical tests used were one-way ANOVA with Sidak’s post-hoc test (a), Tukey’s post-hoc test (d), or Dunnett’s post-hoc test (h,i), two-way ANOVA with Tukey’s post-hoc test (c), and unpaired two-tailed Student’s t test (f,l,n).

Source data

Extended Data Fig. 7 H3D reverses H6D-induced changes in afferent fibers.

a, Discharge frequencies of spontaneously-active fibers (H6D, n = 30; H3D, n = 41). b, Discharge frequencies of AM fibers (H6D, n = 13; H3D, n = 21). *P = 0.0353 (75), 0.0335 (100), **P = 0.0047 (150). c,d, Representative immunofluorescence staining of glabrous hindpaw skin IENFs (c) and ATF3 expression in lumbar DRG neurons (d) in H6D and H3D mice. Scale bars: 25 μm (c; n = 4 mice/group), 50 μm (d; n = 3 mice/group). The representative images supplement Figs. 3h and 3i, respectively. Data are mean ± SEM. Error bars for some data points are within the size of the symbol. Statistical test used was two-way ANOVA with Sidak’s post-hoc test (b).

Source data

Extended Data Fig. 8 Increased LA content in skin of diabetic subjects with painful neuropathy.

a, Chromatogram snapshots of the endogenous LA peak (labeled A) overlaid with the LA-d4 internal control peak (labeled B) for skin biopsy extracts from diabetic and control subjects. Integrated AUC values (a.u.) for each peak are beneath each chromatogram. b-d, Correlation analyses between subject skin LA levels and their respective LANSS scores (b), NPSI scores (c), and hallux vibration detection thresholds (d) (control, n = 12; diabetic, n = 16). Linear regression identified the best-fit line (solid line) with 95% confidence intervals (dotted lines). Inset boxes contain Spearman coefficients (rs) and corresponding P-values. Statistical test used was two-tailed Spearman correlation.

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Table 1. Diet breakdowns. Supplementary Table 2. Clinical data.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Boyd, J.T., LoCoco, P.M., Furr, A.R. et al. Elevated dietary ω-6 polyunsaturated fatty acids induce reversible peripheral nerve dysfunction that exacerbates comorbid pain conditions. Nat Metab 3, 762–773 (2021). https://doi.org/10.1038/s42255-021-00410-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00410-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing