Skip to main content

Role of Nerve Growth Factor in Pain

  • Chapter
Pain Control

Part of the book series: Handbook of Experimental Pharmacology ((HEP,volume 227))

Abstract

Nerve growth factor (NGF) was first identified as a substance that is essential for the development of nociceptive primary neurons and later found to have a role in inflammatory hyperalgesia in adults. Involvement of NGF in conditions with no apparent inflammatory signs has also been demonstrated. In this review we look at the hyperalgesic effects of exogenously injected NGF into different tissues, both human and animal, with special emphasis on the time course of these effects. The roles of NGF in inflammatory and neuropathic conditions as well as cancer pain are then reviewed. The role of NGF in delayed onset muscle soreness is described in more detail than its other roles based on the authors’ recent observations. Acute effects are considered to be peripherally mediated, and accordingly, sensitization of nociceptors by NGF to heat and mechanical stimulation has been reported. Changes in the conductive properties of axons have also been reported. The intracellular mechanisms so far proposed for heat sensitization are direct phosphorylation and membrane trafficking of TRPV1 by TrkA. Little investigation has been done on the mechanism of mechanical sensitization, and it is still unclear whether mechanisms similar to those for heat sensitization work in mechanical sensitization. Long-lasting sensitizing effects are mediated both by changed expression of neuropeptides and ion channels (Na channels, ASIC, TRPV1) in primary afferents and by spinal NMDA receptors. Therapeutic perspectives are briefly discussed at the end of the chapter.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  • Aloe L, Tuveri MA, Carcassi U, Levi-Montalcini R (1992) Nerve growth factor in the synovial fluid of patients with chronic arthritis. Arthritis Rheum 35:351–355

    Article  CAS  PubMed  Google Scholar 

  • Amann R, Schuligoi R, Herzog G, Donnerer J (1996) Intraplantar injection of nerve growth factor into the rat hind paw: local edema and effects on thermal nociceptive threshold. Pain 64:323–329

    Article  CAS  PubMed  Google Scholar 

  • Andersen H, Arendt-Nielsen L, Danneskiold-Samsoe B, Graven-Nielsen T (2006) Pressure pain sensitivity and hardness along human normal and sensitized muscle. Somatosens Mot Res 23:97–109

    Article  PubMed  Google Scholar 

  • Andersen H, Arendt-Nielsen L, Svensson P, Danneskiold-Samsoe B, Graven-Nielsen T (2008) Spatial and temporal aspects of muscle hyperalgesia induced by nerve growth factor in humans. Exp Brain Res 191:371–382

    Article  PubMed  Google Scholar 

  • Andreev NY, Dimitrieva N, Koltzenburg M, Mcmahon SB (1995) Peripheral administration of nerve growth factor in the adult rat produces a thermal hyperalgesia that requires the presence of sympathetic post-ganglionic neurones. Pain 63:109–115

    Article  CAS  PubMed  Google Scholar 

  • Armstrong RB (1984) Mechanisms of exercise-induced delayed onset muscular soreness: a brief review. Med Sci Sports Exerc 16:529–538

    CAS  PubMed  Google Scholar 

  • Bennett DLH, Koltzenburg M, Priestley JV, Shelton DL, Mcmahon SB (1998) Endogenous nerve growth factor regulates the sensitivity of nociceptors in the adult rat. Eur J Neurosci 10:1282–1291

    Article  CAS  PubMed  Google Scholar 

  • Bergmann I, Reiter R, Toyka KV, Koltzenburg M (1998) Nerve growth factor evokes hyperalgesia in mice lacking the low-affinity neurotrophin receptor p75. Neurosci Lett 255:87–90

    Article  CAS  PubMed  Google Scholar 

  • Berry A, Bindocci E, Alleva E (2012) NGF, brain and behavioral plasticity. Neural Plast 2012:784040

    PubMed Central  PubMed  Google Scholar 

  • Bhave G, Hu HJ, Glauner KS, Zhu W, Wang H, Brasier DJ, Oxford GS, Gereau RW (2003) Protein kinase C phosphorylation sensitizes but does not activate the capsaicin receptor transient receptor potential vanilloid 1 (TRPV1). Proc Natl Acad Sci U S A 100:12480–12485

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Bhide AA, Cartwright R, Khullar V, Digesu GA (2013) Biomarkers in overactive bladder. Int Urogynecol J 24:1065–1072

    Article  PubMed  Google Scholar 

  • Bielefeldt K, Ozaki N, Gebhart GF (2003) Role of nerve growth factor in modulation of gastric afferent neurons in the rat. Am J Physiol Gastrointest Liver Physiol 284:G499–G507

    Article  CAS  PubMed  Google Scholar 

  • Bloom AP, Jimenez-Andrade JM, Taylor RN, Castaneda-Corral G, Kaczmarska MJ, Freeman KT, Coughlin KA, Ghilardi JR, Kuskowski MA, Mantyh PW (2011) Breast cancer-induced bone remodeling, skeletal pain, and sprouting of sensory nerve fibers. J Pain 12:698–711

    Article  PubMed Central  PubMed  Google Scholar 

  • Boix F, Rosenborg L, Hilgenfeldt U, Knardahl S (2002) Contraction-related factors affect the concentration of a kallidin-like peptide in rat muscle tissue. J Physiol 544:127–136

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Bonnington JK, McNaughton PA (2003) Signalling pathways involved in the sensitisation of mouse nociceptive neurones by nerve growth factor. J Physiol 551:433–446

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D (1997) The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 389:816–824

    Article  CAS  PubMed  Google Scholar 

  • Caterina MJ, Leffler A, Malmberg AB, Martin WJ, Trafton J, Petersen-Zeitz KR, Kolzenburg M, Basbaum AI, Julius D (2000) Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science 288:306–313

    Article  CAS  PubMed  Google Scholar 

  • Chen TC, Nosaka K, Sacco P (2007) Intensity of eccentric exercise, shift of optimum angle, and the magnitude of repeated-bout effect. J Appl Physiol 102:992–999

    Article  PubMed  Google Scholar 

  • Cheung K, Hume P, Maxwell L (2003) Delayed onset muscle soreness: treatment strategies and performance factors. Sports Med 33:145–164

    Article  PubMed  Google Scholar 

  • Chuang H-H, Prescott ED, Kong H, Shields S, Jordt SE, Basbaum AI, Chao MV, Julius D (2001) Bradykinin and nerve growth factor release the capsaicin receptor from PtdIns(4,5)P2-mediated inhibition. Nature 411:957–962

    Article  CAS  PubMed  Google Scholar 

  • Crameri RM, Aagaard P, Qvortrup K, Langberg H, Olesen J, Kjaer M (2007) Myofibre damage in human skeletal muscle: effects of electrical stimulation versus voluntary contraction. J Physiol 583:365–380

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Crowley C, Spencer SD, Nishimura MC, Chen KS, Pittsmeek S, Armanini MP, Ling LH, Mcmahon SB, Shelton DL, Levinson AD, Phillips HS (1994) Mice lacking nerve growth factor display perinatal loss of sensory and sympathetic neurons yet develop basal forebrain cholinergic neurons. Cell 76:1001–1011

    Article  CAS  PubMed  Google Scholar 

  • Davis JB, Gray J, Gunthorpe MJ, Hatcher JP, Davey PT, Overend P, Harries MH, Latcham J, Clapham C, Atkinson K, Hughes SA, Rance K, Grau E, Harper AJ, Pugh PL, Rogers DC, Bingham S, Randall A, Sheardown SA (2000) Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia. Nature 405:183–187

    Article  CAS  PubMed  Google Scholar 

  • De Col R, Messlinger K, Carr RW (2008) Conduction velocity is regulated by sodium channel inactivation in unmyelinated axons innervating the rat cranial meninges. J Physiol 586:1089–1103

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  • Deising S, Weinkauf B, Blunk J, Obreja O, Schmelz M, Rukwied R (2012) NGF-evoked sensitization of muscle fascia nociceptors in humans. Pain 153:1673–1679

    Article  CAS  PubMed  Google Scholar 

  • Di Castro A, Drew LJ, Wood JN, Cesare P (2006) Modulation of sensory neuron mechanotransduction by PKC- and nerve growth factor-dependent pathways. Proc Natl Acad Sci U S A 103:4699–4704

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  • di Mola FF, Friess H, Zhu ZW, Koliopanos A, Bley T, Di Sebastiano P, Innocenti P, Zimmermann A, Buchler MW (2000) Nerve growth factor and Trk high affinity receptor (TrkA) gene expression in inflammatory bowel disease. Gut 46:670–679

    Article  PubMed  Google Scholar 

  • Djouhri L, Dawbarn D, Robertson A, Newton R, Lawson SN (2001) Time course and nerve growth factor dependence of inflammation-induced alterations in electrophysiological membrane properties in nociceptive primary afferent neurons. J Neurosci 21:8722–8733

    CAS  PubMed  Google Scholar 

  • Donnerer J, Schuligoi R, Stein C (1992) Increased content and transport of substance P and calcitonin gene-related peptide in sensory nerves innervating inflamed tissue: evidence for a regulatory function of nerve growth factor in vivo. Neuroscience 49:693–698

    Article  CAS  PubMed  Google Scholar 

  • Donnerer J, Schuligoi R, Stein C, Amann R (1993) Upregulation, release and axonal transport of substance-P and calcitonin gene-related peptide in adjuvant inflammation and regulatory function of nerve growth factor. Regul Pept 46:150–154

    CAS  PubMed  Google Scholar 

  • Donnerer J, Liebmann I, Schicho R (2005) Differential regulation of 3-beta-hydroxysteroid dehydrogenase and vanilloid receptor TRPV1 mRNA in sensory neurons by capsaicin and NGF. Pharmacology 73:97–101

    Article  CAS  PubMed  Google Scholar 

  • Dyck PJ, Peroutka S, Rask C, Burton E, Baker MK, Lehman KA, Gillen DA, Hokanson JL, O’Brien PC (1997) Intradermal recombinant human nerve growth factor induces pressure allodynia and lowered heat-pain threshold in humans. Neurology 48:501–505

    Article  CAS  PubMed  Google Scholar 

  • Eibl JK, Strasser BC, Ross GM (2012) Structural, biological, and pharmacological strategies for the inhibition of nerve growth factor. Neurochem Int 61:1266–1275

    Article  CAS  PubMed  Google Scholar 

  • Einarsdottir E, Carlsson A, Minde J, Toolanen G, Svensson O, Solders G, Holmgren G, Holmberg D, Holmberg M (2004) A mutation in the nerve growth factor beta gene (NGFB) causes loss of pain perception. Hum Mol Genet 13:799–805

    Article  CAS  PubMed  Google Scholar 

  • Fjell J, Cummins TR, Dib-Hajj SD, Fried K, Black JA, Waxman SG (1999) Differential role of GDNF and NGF in the maintenance of two TTX-resistant sodium channels in adult DRG neurons. Mol Brain Res 67:267–282

    Article  CAS  PubMed  Google Scholar 

  • Freemont AJ, Watkins A, Le MC, Baird P, Jeziorska M, Knight MT, Ross ER, O’Brien JP, Hoyland JA (2002) Nerve growth factor expression and innervation of the painful intervertebral disc. J Pathol 197:286–292

    Article  CAS  PubMed  Google Scholar 

  • Fujii Y, Ozaki N, Taguchi T, Mizumura K, Sugiura Y (2008) TRP channels and ASICs mediate mechanical hyperalgesia in models of inflammatory muscle pain and delayed onset muscle soreness. Pain 140:292–304

    Article  CAS  PubMed  Google Scholar 

  • Fukuoka T, Kondo E, Dai Y, Hashimoto N, Noguchi K (2001) Brain-derived neurotrophic factor increases in the uninjured dorsal root ganglion neurons in selective spinal nerve ligation model. J Neurosci 21:4891–4900

    CAS  PubMed  Google Scholar 

  • Ghilardi JR, Freeman KT, Jimenez-Andrade JM, Mantyh WG, Bloom AP, Bouhana KS, Trollinger D, Winkler J, Lee P, Andrews SW, Kuskowski MA, Mantyh PW (2011) Sustained blockade of neurotrophin receptors TrkA, TrkB and TrkC reduces non-malignant skeletal pain but not the maintenance of sensory and sympathetic nerve fibers. Bone 48:389–398

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Giovengo SL, Russell IJ, Larson AA (1999) Increased concentrations of nerve growth factor in cerebrospinal fluid of patients with fibromyalgia. J Rheumatol 26:1564–1569

    CAS  PubMed  Google Scholar 

  • Guo TZ, Offley SC, Boyd EA, Jacobs CR, Kingery WS (2004) Substance P signaling contributes to the vascular and nociceptive abnormalities observed in a tibial fracture rat model of complex regional pain syndrome type I. Pain 108:95–107

    Article  CAS  PubMed  Google Scholar 

  • Halliday DA, Zettler C, Rush RA, Scicchitano R, McNeil JD (1998) Elevated nerve growth factor levels in the synovial fluid of patients with inflammatory joint disease. Neurochem Res 23:919–922

    Article  CAS  PubMed  Google Scholar 

  • Halvorson KG, Kubota K, Sevcik MA, Lindsay TH, Sotillo JE, Ghilardi JR, Rosol TJ, Boustany L, Shelton DL, Mantyh PW (2005) A blocking antibody to nerve growth factor attenuates skeletal pain induced by prostate tumor cells growing in bone. Cancer Res 65:9426–9435

    Article  CAS  PubMed  Google Scholar 

  • Hayashi K, Shiozawa S, Ozaki N, Mizumura K, Graven-Nielsen T (2013) Repeated intramuscular injections of nerve growth factor induced progressive muscle hyperalgesia, facilitated temporal summation and expanded pain areas. Pain 154:2344–2352

    Article  CAS  PubMed  Google Scholar 

  • Hirth M, Rukwied R, Gromann A, Turnquist B, Weinkauf B, Francke K, Albrecht P, Rice F, Hagglof B, Ringkamp M, Engelhardt M, Schultz C, Schmelz M, Obreja O (2013) Nerve growth factor induces sensitization of nociceptors without evidence for increased intraepidermal nerve fiber density. Pain 154:2500–2511

    Article  CAS  PubMed  Google Scholar 

  • Hoheisel U, Unger T, Mense S (2005) Excitatory and modulatory effects of inflammatory cytokines and neurotrophins on mechanosensitive group IV muscle afferents in the rat. Pain 114:168–176

    Article  CAS  PubMed  Google Scholar 

  • Hoheisel U, Taguchi T, Treede RD, Mense S (2011) Nociceptive input from the rat thoracolumbar fascia to lumbar dorsal horn neurones. Eur J Pain 15:810–815

    Article  PubMed  Google Scholar 

  • Hoheisel U, Taguchi T, Mense S (2012) Nociception: the thoracolumbar fascia as a sensory organ. In: Schleip R, Findley TW, Chaitow L, Huijing PA (eds) Fascia: the tensional network of the human body: the science and clinical applications in manual and movement therapy. Churchill Livingstone/Elsevier, Edinburgh, pp 95–101

    Google Scholar 

  • Iannone F, De BC, Dell’Accio F, Covelli M, Patella V, Lo BG, Lapadula G (2002) Increased expression of nerve growth factor (NGF) and high affinity NGF receptor (p140 TrkA) in human osteoarthritic chondrocytes. Rheumatology 41:1413–1418

    Article  CAS  PubMed  Google Scholar 

  • Indo Y, Tsuruta M, Hayashida Y, Karim MA, Ohta K, Kawano T, Mitsubuchi H, Tonoki H, Awaya Y, Matsuda I (1996) Mutations in the TRKA/NGF receptor gene in patients with congenital insensitivity to pain with anhidrosis. Nat Genet 13:485–488

    Article  CAS  PubMed  Google Scholar 

  • Itoh K, Okada K, Kawakita K (2004) A proposed experimental model of myofascial trigger points in human muscle after slow eccentric exercise. Acupunct Med 22:2–12

    Article  PubMed  Google Scholar 

  • Iwakura N, Ohtori S, Orita S, Yamashita M, Takahashi K, Kuniyoshi K (2010) Role of low-affinity nerve growth factor receptor inhibitory antibody in reducing pain behavior and calcitonin gene-related Peptide expression in a rat model of wrist joint inflammatory pain. J Hand Surg Am 35:267–273

    Article  PubMed  Google Scholar 

  • Ji R, Samad T, Jin S, Schmoll R, Woolf C (2002) p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 36:57–68

    Article  CAS  PubMed  Google Scholar 

  • Jiang YH, Peng CH, Liu HT, Kuo HC (2013) Increased pro-inflammatory cytokines, C-reactive protein and nerve growth factor expressions in serum of patients with interstitial cystitis/bladder pain syndrome. PLoS One 8:e76779

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Kawakita K, Itoh K, Okada K (2008) Experimental model of trigger points using eccentric exercise. J Musculoskelet Pain 16:29–35

    Article  Google Scholar 

  • Khodorova A, Nicol GD, Strichartz G (2013) The p75(NTR) signaling cascade mediates mechanical hyperalgesia induced by nerve growth factor injected into the rat hind paw. Neuroscience 254:312–323

    Article  CAS  PubMed  Google Scholar 

  • Kim JC, Park EY, Seo SI, Park YH, Hwang TK (2006) Nerve growth factor and prostaglandins in the urine of female patients with overactive bladder. J Urol 175:1773–1776

    Article  CAS  PubMed  Google Scholar 

  • Kivitz AJ, Gimbel JS, Bramson C, Nemeth MA, Keller DS, Brown MT, West CR, Verburg KM (2013) Efficacy and safety of tanezumab versus naproxen in the treatment of chronic low back pain. Pain 154:1009–1021

    Article  CAS  PubMed  Google Scholar 

  • Koltzenburg M, Bennett DL, Shelton DL, Mcmahon SB (1999) Neutralization of endogenous NGF prevents the sensitization of nociceptors supplying inflamed skin. Eur J Neurosci 11:1698–1704

    Article  CAS  PubMed  Google Scholar 

  • Lamb K, Gebhart GF, Bielefeldt K (2004) Increased nerve growth factor expression triggers bladder overactivity. J Pain 5:150–156

    Article  CAS  PubMed  Google Scholar 

  • Leslie TA, Emson PC, Dowd PM, Woolf CJ (1995) Nerve growth factor contributes to the up-regulation of growth- associated protein 43 and preprotachykinin A messenger RNAs in primary sensory neurons following peripheral inflammation. Neuroscience 67:753–761

    Article  CAS  PubMed  Google Scholar 

  • Lewin GR, Nykjaer A (2014) Pro-neurotrophins, sortilin, and nociception. Eur J Neurosci 39:363–374

    Article  PubMed Central  PubMed  Google Scholar 

  • Lewin GR, Winter J, Mcmahon SB (1992) Regulation of afferent connectivity in the adult spinal cord by nerve growth factor. Eur J Neurosci 4:700–707

    Article  CAS  PubMed  Google Scholar 

  • Lewin GR, Ritter AM, Mendell LM (1993) Nerve growth factor-induced hyperalgesia in the neonatal and adult rat. J Neurosci 13:2136–2148

    CAS  PubMed  Google Scholar 

  • Lewin GR, Rueff A, Mendell LM (1994) Peripheral and central mechanisms of NGF-induced hyperalgesia. Eur J Neurosci 6:1903–1912

    Article  CAS  PubMed  Google Scholar 

  • Liu HT, Chancellor MB, Kuo HC (2009) Decrease of urinary nerve growth factor levels after antimuscarinic therapy in patients with overactive bladder. BJU Int 103:1668–1672

    Article  CAS  PubMed  Google Scholar 

  • Liu HT, Jiang YH, Kuo HC (2013) Increased serum adipokines implicate chronic inflammation in the pathogenesis of overactive bladder syndrome refractory to antimuscarinic therapy. PLoS One 8:e76706

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Malik-Hall M, Dina OA, Levine JD (2005) Primary afferent nociceptor mechanisms mediating NGF-induced mechanical hyperalgesia. Eur J Neurosci 21:3387–3394

    Article  PubMed  Google Scholar 

  • Mamet J, Baron A, Lazdunski M, Voilley N (2002) Proinflammatory mediators, stimulators of sensory neuron excitability via the expression of acid-sensing ion channels. J Neurosci 22:10662–10670

    CAS  PubMed  Google Scholar 

  • Mann MK, Dong XD, Svensson P, Cairns BE (2006) Influence of intramuscular nerve growth factor injection on the response properties of rat masseter muscle afferent fibers. J Orofac Pain 20:325–336

    PubMed  Google Scholar 

  • Matsuura Y, Iwakura N, Ohtori S, Suzuki T, Kuniyoshi K, Murakami K, Hiwatari R, Hashimoto K, Okamoto S, Shibayama M, Kobayashi T, Ogawa Y, Sukegawa K, Takahashi K (2013) The effect of Anti-NGF receptor (p75 Neurotrophin Receptor) antibodies on nociceptive behavior and activation of spinal microglia in the rat brachial plexus avulsion model. Spine (Phila Pa 1976) 38:E332–E338

    Article  Google Scholar 

  • Mcmahon SB, Bennett DL, Priestley JV, Shelton DL (1995) The biological effects of endogenous nerve growth factor on adult sensory neurons revealed by a trkA-IgG fusion molecule. Nat Med 1:774–780

    Article  CAS  PubMed  Google Scholar 

  • McNamee KE, Burleigh A, Gompels LL, Feldmann M, Allen SJ, Williams RO, Dawbarn D, Vincent TL, Inglis JJ (2010) Treatment of murine osteoarthritis with TrkAd5 reveals a pivotal role for nerve growth factor in non-inflammatory joint pain. Pain 149:386–392

    Article  CAS  PubMed  Google Scholar 

  • Murase S, Terazawa E, Queme F, Ota H, Matsuda T, Hirate K, Kozaki Y, Katanosaka K, Taguchi T, Mizumura K (2010) Bradykinin and nerve growth factor play pivotal roles in muscular mechanical hyperalgesia after exercise (delayed onset muscle soreness). J Neurosci 30:3752–3761

    Article  CAS  PubMed  Google Scholar 

  • Murase S, Yamanaka Y, Kanda H, Mizumura K (2012) COX-2, nerve growth factor (NGF) and glial cell-derived neurotrophic factor (GDNF), which play pivotal roles in delayed onset muscle soreness (DOMS), are produced by exercised skeletal muscle. J Physiol Sci 62(Suppl):S179

    Google Scholar 

  • Murase S, Terazawa E, Hirate K, Yamanaka H, Kanda H, Noguchi K, Ota H, Queme F, Taguchi T, Mizumura K (2013) Upregulated glial cell line-derived neurotrophic factor through cyclooxygenase-2 activation in the muscle is required for mechanical hyperalgesia after exercise in rats. J Physiol 591:3035–3048

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Nicol GD, Vasko MR (2007) Unraveling the story of NGF-mediated sensitization of nociceptive sensory neurons: ON or OFF the Trks? Mol Interv 7:26–41

    Article  CAS  PubMed  Google Scholar 

  • Nishigami T, Osako Y, Ikeuchi M, Yuri K, Ushida T (2013) Development of heat hyperalgesia and changes of TRPV1 and NGF expression in rat dorsal root ganglion following joint immobilization. Physiol Res 62:215–219

    CAS  PubMed  Google Scholar 

  • Oddiah D, Anand P, Mcmahon SB, Rattray M (1998) Rapid increase of NGF, BDNF and NT-3 mRNAs in inflamed bladder. Neuroreport 9:1455–1458

    Article  CAS  PubMed  Google Scholar 

  • Ohmichi Y, Sato J, Ohmichi M, Sakurai H, Yoshimoto T, Morimoto A, Hashimoto T, Eguchi K, Nishihara M, Arai YC, Ohishi H, Asamoto K, Ushida T, Nakano T, Kumazawa T (2012) Two-week cast immobilization induced chronic widespread hyperalgesia in rats. Eur J Pain 16:338–348

    Article  CAS  PubMed  Google Scholar 

  • Ota H, Katanosaka K, Murase S, Kashio M, Tominaga M, Mizumura K (2013) TRPV1 and TRPV4 play pivotal roles in delayed onset muscle soreness. PLoS One 8:e65751

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Pantano F, Zoccoli A, Iuliani M, Lanzetta G, Vincenzi B, Tonini G, Santini D (2011) New targets, new drugs for metastatic bone pain: a new philosophy. Expert Opin Emerg Drugs 16:403–405

    Article  CAS  PubMed  Google Scholar 

  • Petty BG, Cornblath DR, Adornato BT, Chaudhry V, Flexner C, Wachsman M, Sinicropi D, Burton LE, Peroutka SJ (1994) The effect of systemically administered recombinant human nerve growth factor in healthy human subjects. Ann Neurol 36:244–246

    Article  CAS  PubMed  Google Scholar 

  • Queme F, Taguchi T, Mizumura K, Graven-Nielsen T (2013) Muscular heat and mechanical pain sensitivity after lengthening contractions in humans and animals. J Pain 14:1425–1436

    Article  PubMed  Google Scholar 

  • Ritter AM, Lewin GR, Kremer NE, Mendell LM (1991) Requirement for nerve growth factor in the development of myelinated nociceptors in vivo. Nature 350:500–502

    Article  CAS  PubMed  Google Scholar 

  • Rukwied R, Mayer A, Kluschina O, Obreja O, Schley M, Schmelz M (2010) NGF induces non-inflammatory localized and lasting mechanical and thermal hypersensitivity in human skin. Pain 148:407–413

    Article  CAS  PubMed  Google Scholar 

  • Sanga P, Katz N, Polverejan E, Wang S, Kelly KM, Haeussler J, Thipphawong J (2013) Efficacy, safety, and tolerability of fulranumab, an anti-nerve growth factor antibody, in the treatment of patients with moderate to severe osteoarthritis pain. Pain 154:1910–1919

    Article  CAS  PubMed  Google Scholar 

  • Sarchielli P, Mancini ML, Floridi A, Coppola F, Rossi C, Nardi K, Acciarresi M, Pini LA, Calabresi P (2007) Increased levels of neurotrophins are not specific for chronic migraine: evidence from primary fibromyalgia syndrome. J Pain 8:737–745

    Article  CAS  PubMed  Google Scholar 

  • Schmieg N, Menendez G, Schiavo G, Terenzio M (2014) Signalling endosomes in axonal transport: travel updates on the molecular highway. Semin Cell Dev Biol 27:32–43. doi:10.1016/j.semcdb.2013.10.004

    Article  CAS  PubMed  Google Scholar 

  • Sekino Y, Nakano J, Hamaue Y, Chuganji S, Sakamoto J, Yoshimura T, Origuchi T, Okita M (2014) Sensory hyperinnervation and increase in NGF, TRPV1 and P2X3 expression in the epidermis following cast immobilization in rats. Eur J Pain 18:639–648

    Article  CAS  PubMed  Google Scholar 

  • Shu X, Mendell LM (1999) Nerve growth factor acutely sensitizes the response of adult rat sensory neurons to capsaicin. Neurosci Lett 274:159–162

    Article  CAS  PubMed  Google Scholar 

  • Shu X, Mendell LM (2001) Acute sensitization by NGF of the response of small-diameter sensory neurons to capsaicin. J Neurophysiol 86:2931–2938

    CAS  PubMed  Google Scholar 

  • Smith LL (1991) Acute inflammation—the underlying mechanism in delayed onset muscle soreness. Med Sci Sports Exerc 23:542–551

    CAS  PubMed  Google Scholar 

  • Stein AT, Ufret-Vincenty CA, Hua L, Santana LF, Gordon SE (2006) Phosphoinositide 3-kinase binds to TRPV1 and mediates NGF-stimulated TRPV1 trafficking to the plasma membrane. J Gen Physiol 128:509–522

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Svensson P, Cairns BE, Wang K, Arendt-Nielsen L (2003) Injection of nerve growth factor into human masseter muscle evokes long-lasting mechanical allodynia and hyperalgesia. Pain 104:241–247

    Article  CAS  PubMed  Google Scholar 

  • Taguchi T, Matsuda T, Tamura R, Sato J, Mizumura K (2005a) Muscular mechanical hyperalgesia revealed by behavioural pain test and c-Fos expression in the spinal dorsal horn after eccentric contraction in rats. J Physiol 564:259–268

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Taguchi T, Sato J, Mizumura K (2005b) Augmented mechanical response of muscle thin-fiber sensory receptors recorded from rat muscle-nerve preparations in vitro after eccentric contraction. J Neurophysiol 94:2822–2831

    Article  PubMed  Google Scholar 

  • Taguchi T, Hoheisel U, Mense S (2008a) Dorsal horn neurons having input from low back structures in rats. Pain 138:119–129

    Article  PubMed  Google Scholar 

  • Taguchi T, Hoheisel U, Mense S (2008b) Neuroanatomy and electrophysiology of low back pain: experiments on rats. Aktuelle Urol 34:472–477

    Article  Google Scholar 

  • Taguchi T, Yasui M, Kubo A, Abe M, Kiyama H, Yamanaka A, Mizumura K (2013) Nociception originating from the crural fascia in rats. Pain 154:1103–1114

    Article  CAS  PubMed  Google Scholar 

  • Urai H, Murase S, Mizumura K (2012) Decreased nerve growth factor upregulation is a mechanism for reduced mechanical hyperalgesia after the second bout of exercise in rats. Scand J Med Sci Sports 23:e96–e101

    Article  PubMed  Google Scholar 

  • Wild KD, Bian D, Zhu D, Davis J, Bannon AW, Zhang TJ, Louis JC (2007) Antibodies to nerve growth factor reverse established tactile allodynia in rodent models of neuropathic pain without tolerance. J Pharmacol Exp Ther 322:282–287

    Article  CAS  PubMed  Google Scholar 

  • Woolf CJ, Safieh-Garabedian B, Ma QP, Crilly P, Winter J (1994) Nerve growth factor contributes to the generation of inflammatory sensory hypersensitivity. Neuroscience 62:327–331

    Article  CAS  PubMed  Google Scholar 

  • Woolf CJ, Ma QP, Allchorne A, Poole S (1996) Peripheral cell types contributing to the hyperalgesic action of nerve growth factor in inflammation. J Neurosci 16:2716–2723

    CAS  PubMed  Google Scholar 

  • Xue Q, Jong B, Chen T, Schumacher MA (2007) Transcription of rat TRPV1 utilizes a dual promoter system that is positively regulated by nerve growth factor. J Neurochem 101:212–222

    Article  CAS  PubMed  Google Scholar 

  • Ye Y, Dang D, Zhang J, Viet CT, Lam DK, Dolan JC, Gibbs JL, Schmidt BL (2011) Nerve growth factor links oral cancer progression, pain, and cachexia. Mol Cancer Ther 10:1667–1676

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Zhang X, Huang J, McNaughton PA (2005) NGF rapidly increases membrane expression of TRPV1 heat-gated ion channels. EMBO J 24:4211–4223

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Zhang YH, Kays J, Hodgdon KE, Sacktor TC, Nicol GD (2012) Nerve growth factor enhances the excitability of rat sensory neurons through activation of the atypical protein kinase C isoform, PKMzeta. J Neurophysiol 107:315–335

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Zhu W, Oxford GS (2007) Phosphoinositide-3-kinase and mitogen activated protein kinase signaling pathways mediate acute NGF sensitization of TRPV1. Mol Cell Neurosci 34:689–700

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  • Zhu W, Galoyan SM, Petruska JC, Oxford GS, Mendell LM (2004) A developmental switch in acute sensitization of small dorsal root ganglion (DRG) neurons to capsaicin or noxious heating by NGF. J Neurophysiol 92:3148–3152

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kazue Mizumura .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2015 Springer-Verlag Berlin Heidelberg

About this chapter

Cite this chapter

Mizumura, K., Murase, S. (2015). Role of Nerve Growth Factor in Pain. In: Schaible, HG. (eds) Pain Control. Handbook of Experimental Pharmacology, vol 227. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-662-46450-2_4

Download citation

Publish with us

Policies and ethics