Dry needling at myofascial trigger spots of rabbit skeletal muscles modulates the biochemicals associated with pain, inflammation, and hypoxia

Yueh-Ling Hsieh, Shun-An Yang, Chen-Chia Yang, Li-Wei Chou, Yueh-Ling Hsieh, Shun-An Yang, Chen-Chia Yang, Li-Wei Chou

Abstract

Background and Purpose. Dry needling is an effective therapy for the treatment of pain associated with myofascial trigger point (MTrP). However, the biochemical effects of dry needling that are associated with pain, inflammation, and hypoxia are unclear. This study investigated the activities of β-endorphin, substance P, TNF-α, COX-2, HIF-1α, iNOS, and VEGF after different dosages of dry needling at the myofascial trigger spots (MTrSs) of a skeletal muscle in rabbit. Materials and Methods. Dry needling was performed either with one dosage (1D) or five dosages (5D) into the biceps femoris with MTrSs in New Zealand rabbits. Biceps femoris, serum, and dorsal root ganglion (DRG) were sampled immediately and 5 d after dry needling for β-endorphin, substance P, TNF-α, COX-2, HIF-1α, iNOS, and VEGF immunoassays. Results. The 1D treatment enhanced the β-endorphin levels in the biceps femoris and serum and reduced substance P in the biceps femoris and DRG. The 5D treatment reversed these effects and was accompanied by increase of TNF-α, COX-2, HIF-1α, iNOS, and VEGF production in the biceps femoris. Moreover, the higher levels of these biochemicals were still maintained 5 d after treatment. Conclusion. Dry needling at the MTrSs modulates various biochemicals associated with pain, inflammation, and hypoxia in a dose-dependent manner.

Figures

Figure 1
Figure 1
Flow chart for the animal study. Abbreviations: 1D, one-dosage dry needling; 5D, five-dosage dry needling; BF, biceps femoris; COX-2, cyclooxygenase-2; DRG, dorsal root ganglion; End, β-endorphin; HIF-1α, hypoxia-inducible factor-1α; iNOS, inducible isoform of nitric oxide synthases; MTrS, myofascial trigger spot; SP, substance P; TNF-α, tumor necrosis factor-α; VEGF, vascular endothelial growth factor; s1D, sham one-dosage dry needling; s5D, sham five-dosage dry needling.
Figure 2
Figure 2
Morphological findings of representative skeletal muscles with nontaut and taut bands. (a) Biceps femoris with a nontaut band; (b) Biceps femoris with a taut band (H&E staining, scale bar = 5 μm).
Figure 3
Figure 3
Effects of one- and five-dosage (1D, 5D) dry needling on β-endorphin expression in the needling-treated muscle and serum. The levels of β-endorphin in the muscle and serum obtained by (a) Western blot analysis and (b) ELISA. A representative Western blot image of a muscle tissue is shown (upper panel of (a)). The quantification of the protein levels is expressed as mean ± SD. *Indicates a significant difference (P < 0.05) between the sham groups (s1D and s5D). #Represents the significant difference (P < 0.05) between the 1D and 5D groups. DN: dry needling.
Figure 4
Figure 4
Effects of one- and five-dosage dry needling (1D, 5D) on substance P expression in a (a) needling-treated muscle and (b) DRG. Representative Western blot images are presented on the upper panels of (a) and (b). The quantification of the protein levels is expressed as mean ± SD. *Indicates the significant difference (P < 0.05) between the sham groups (s1D and s5D). #Represents the significant difference (P < 0.05) between the 1D and 5D groups. DN: dry needling.
Figure 5
Figure 5
Effects of one- and five-dosage dry needling (1D, 5D) on iNOS, HIF-1α, COX-2, and VEGF expressions in a needling-treated muscle. (a) Representative Western blot images. The quantification of the protein levels for (b) iNOS, (c) HIF-1α, (d) COX-2, and (e) VEGF. Values are expressed as mean ± SD. *Indicates the significant difference (P < 0.05) between the sham groups (s1D and s5D). #Represents the significant difference (P < 0.05) between the 1D and 5D groups. DN: dry needling.
Figure 6
Figure 6
The variation in TNF-α after one- and five-dosage dry needling (1D, 5D) in the biceps femoris with a taut band in the experimental (1D, 5D) and sham groups (s1D, s5D). Representative photomicrographs indicate the immunohistochemical labeling for TNF-α in the muscle immediately after dry needling: (a) s1D, (b) 1D, (c) s5D, and (d) 5D, as well as 5 d after dry needling: (e) s1D, (f) 1D, (g) s5D, and (h) 5D. Histograms indicate the quantitative analysis of (i) TNF-LI cells and (j) protein levels of TNF-α by applying ELISA in the serum. *Indicates the significant difference (P < 0.05) between the sham groups (s1D and s5D). #Represents the significant difference (P < 0.05) between the 1D and 5D groups. DN: dry needling. (scale bar = 5 μm).

References

    1. Gerwin RD. Myofascial aspects of low back pain. Neurosurgery clinics of North America. 1991;2(4):761–784.
    1. Hong CZ, Simons DG. Pathophysiologic and electrophysiologic mechanisms of myofascial trigger points. Archives of Physical Medicine and Rehabilitation. 1998;79(7):863–872.
    1. Simons DG, Travell JG, Simons LS. Travell and Simons’ Myofascial Pain and Dysfunction: The Trigger Point Manual. 2nd edition. Baltimore, Pa, USA: Lippincott Williams & Wilkins; 1999.
    1. Hong CZ. Research on myofascial pain syndrome. Critical Reviews in Physical and Rehabilitation Medicine. 2008;20(4):343–366.
    1. Jarrell J. Myofascial pain in the adolescent. Current Opinion in Obstetrics and Gynecology. 2010;22(5):393–398.
    1. Malanga GA, Colon EJC. Myofascial low back pain: a review. Physical Medicine and Rehabilitation Clinics of North America. 2010;21(4):711–724.
    1. Mense S, Simons DG. Muscle Pain: Understanding Its Nature, Diagnosis, and Treatment. Lippincott Williams & Wilkins edition. Philadelphia, Pa, USA: 2001.
    1. Fricton JR, Auvinen MD, Dykstra D, Schiffman E. Myofascial pain syndrome. Electromyographic changes associated with local twitch response. Archives of Physical Medicine and Rehabilitation. 1985;66(5):314–317.
    1. Shah JP, Danoff JV, Desai MJ, et al. Biochemicals associated with pain and inflammation are elevated in sites near to and remote from active myofascial trigger points. Archives of Physical Medicine and Rehabilitation. 2008;89(1):16–23.
    1. Lembeck F, Donnerer J. Opioid control of the function of primary afferent substance P fibres. European Journal of Pharmacology. 1985;114(3):241–246.
    1. Shah JP, Gilliams EA. Uncovering the biochemical milieu of myofascial trigger points using in vivo microdialysis: an application of muscle pain concepts to myofascial pain syndrome. Journal of Bodywork and Movement Therapies. 2008;12(4):371–384.
    1. Hsieh YL, Hong CZ. Laser therapy for myofascial pain. Critical Reviews in Physical and Rehabilitation Medicine. 2010;22(1–4):241–278.
    1. Tang K, Breen EC, Wagner H, Brutsaert TD, Gassmann M, Wagner PD. HIF and VEGF relationships in response to hypoxia and sciatic nerve stimulation in rat gastrocnemius. Respiratory Physiology and Neurobiology. 2004;144(1):71–80.
    1. Hong CZ. New trends in myofascial pain syndrome. Chinese Medical Journal. 2002;65(11):501–512.
    1. Huang YT, Lin SY, Neoh CA, Wang KY, Jean YH, Shi HY. Dry needling for myofascial pain: prognostic factors. Journal of Alternative and Complementary Medicine. 2011;17(8):755–762.
    1. Kalichman L, Vulfsons S. Dry needling in the management of musculoskeletal pain. Journal of the American Board of Family Medicine. 2010;23(5):640–646.
    1. Hsieh YL, Kao MJ, Kuan TS, Chen SM, Chen JT, Hong CZ. Dry needling to a key myofascial trigger point may reduce the irritability of satellite MTrPs. American Journal of Physical Medicine and Rehabilitation. 2007;86(5):397–403.
    1. Cummings TM, White AR. Needling therapies in the management of myofascial trigger point pain: a systematic review. Archives of Physical Medicine and Rehabilitation. 2001;82(7):986–992.
    1. Hong CZ. Lidocaine injection versus dry needling to myofascial trigger point: the importance of the local twitch response. American Journal of Physical Medicine and Rehabilitation. 1994;73(4):256–263.
    1. Tough EA, White AR, Cummings TM, Richards SH, Campbell JL. Acupuncture and dry needling in the management of myofascial trigger point pain: a systematic review and meta-analysis of randomised controlled trials. European Journal of Pain. 2009;13(1):3–10.
    1. Zimmermann M. Ethical considerations in relation to pain in animal experimentation. Acta Physiologica Scandinavica. 1986;128(554):221–233.
    1. Zimmermann M. Ethical guidelines for investigations of experimental pain in conscious animals. Pain. 1983;16(2):109–110.
    1. Chen KH, Hong CZ, Kuo FC, Hsu HC, Hsieh YL. Electrophysiologic effects of a therapeutic laser on myofascial trigger spots of rabbit skeletal muscles. American Journal of Physical Medicine and Rehabilitation. 2008;87(12):1006–1014.
    1. Chen KH, Hong CZ, Hsu HC, Wu SK, Kuo FC, Hsieh YL. Dose-dependent and ceiling effects of therapeutic laser on myofascial trigger spots in rabbit skeletal muscles. Journal of Musculoskeletal Pain. 2010;18(3):235–245.
    1. Hsieh YL, Chou LW, Joe YS, Hong CZ. Spinal cord mechanism involving the remote effects of dry needling on the irritability of myofascial trigger spots in rabbit skeletal muscle. Archives of Physical Medicine and Rehabilitation. 2011;92(7):1098–1105.
    1. Hong CZ, Torigoe Y. Electrophysiological characteristics of localized twitch responses in responsive taut bands of rabbit skeletal muscle fibers. Journal of Musculoskeletal Pain. 1994;2(2):17–43.
    1. Simons DG, Hong CZ, Simons LS. Prevalence of spontaneous electrical activity at trigger spots and at control sites in rabbit skeletal muscle. Journal of Musculoskeletal Pain. 1995;3(1):35–48.
    1. Simons DG, Hong CZ, Simons LS. Endplate potentials are common to midfiber myofacial trigger points. American Journal of Physical Medicine and Rehabilitation. 2002;81(3):212–222.
    1. Kuan TS, Chen JT, Chen SM, Chien CH, Hong CZ. Effect of botulinum toxin on endplate noise in myofascial trigger spots of rabbit skeletal muscle. American Journal of Physical Medicine and Rehabilitation. 2002;81(7):512–520.
    1. Kuan TS, Hong CZ, Chen JT, Chen SM, Chien CH. The spinal cord connections of the myofascial trigger spots. European Journal of Pain. 2007;11(6):624–634.
    1. Wood PL. Animal models in analgesic testing. In: Kuhar MJ, Pasternak GW, editors. Central Nervous System Pharmacology. Analgesics: Neurochemical, Behavioral and Clinical Perspective. New York, NY, USA: Raven Press; 1984. pp. 175–194.
    1. Chou LW, Hsieh YL, Kao MJ, Hong CZ. Remote influences of acupuncture on the pain intensity and the amplitude changes of endplate noise in the myofascial trigger point of the upper trapezius muscle. Archives of Physical Medicine and Rehabilitation. 2009;90(6):905–912.
    1. Sekido R, Ishimaru K, Sakita M. Differences of electroacupuncture-induced analgesic effect in normal and inflammatory conditions in rats. American Journal of Chinese Medicine. 2003;31(6):955–965.
    1. Taguchi R, Taguchi T, Kitakoji H. Involvement of peripheral opioid receptors in electroacupuncture analgesia for carrageenan-induced hyperalgesia. Brain Research. 2010;1355:97–103.
    1. Yang ES, Li PW, Nilius B, Li G. Ancient Chinese medicine and mechanistic evidence of acupuncture physiology. Pflügers Archiv. 2011;462(5):645–653.
    1. Lee HJ, Lee JH, Lee EO, et al. Substance P and beta endorphin mediate electroacupuncture induced analgesic activity in mouse cancer pain model. Acupuncture & Electro-Therapeutics Research. 2009;34(1-2):27–40.
    1. McMillan AS, Nolan A, Kelly PJ. The efficacy of dry needling and procaine in the treatment of myofascial pain in the jaw muscles. Journal of Orofacial Pain. 1997;11(4):307–314.
    1. Garvey TA, Marks MR, Wiesel SW. A prospective, randomized, double-blind evaluation of trigger-point injection therapy for low-back pain. Spine. 1989;14(9):962–964.
    1. Chiang J, Shen YC, Wang YH, et al. Honokiol protects rats against eccentric exercise-induced skeletal muscle damage by inhibiting NF-κB induced oxidative stress and inflammation. European Journal of Pharmacology. 2009;610(1–3):119–127.
    1. Inoue A, Ikoma K, Morioka N, et al. Interleukin-1β induces substance P release from primary afferent neurons through the cyclooxygenase-2 system. Journal of Neurochemistry. 1999;73(5):2206–2213.
    1. Liou JT, Liu FC, Mao CC, Lai YS, Day YJ. Inflammation confers dual effects on nociceptive processing in chronic neuropathic pain model. Anesthesiology. 2011;114(3):660–672.
    1. Feldreich A, Ernberg M, Lund B, Rosen A. Increased beta-endorphin levels and generalized decreased pain thresholds in patients with limited jaw opening and movement-evoked pain from the temporomandibular joint. Journal of Oral and Maxillofacial Surgery. 2012;70(3):547–556.
    1. Milkiewicz M, Haas TL. Effect of mechanical stretch on HIF-1α and MMP-2 expression in capillaries isolated from overloaded skeletal muscles: laser capture microdissection study. American Journal of Physiology. 2005;289(3):H1315–H1320.
    1. Fang Li Q, Xu H, Sun Y, Hu R, Jiang H. Induction of inducible nitric oxide synthase by isoflurane post-conditioning via hypoxia inducible factor-1α during tolerance against ischemic neuronal injury. Brain Research. 2012;1451:1–9.
    1. Loor G, Schumacker PT. Role of hypoxia-inducible factor in cell survival during myocardial ischemia-reperfusion. Cell Death and Differentiation. 2008;15(4):686–690.
    1. Bergeron M, Yu AY, Solway KE, Semenza GL, Sharp FR. Induction of hypoxia-inducible factor-1 (HIF-1) and its target genes following focal ischaemia in rat brain. European Journal of Neuroscience. 1999;11(12):4159–4170.
    1. Rosendal L, Larsson B, Kristiansen J, et al. Increase in muscle nociceptive substances and anaerobic metabolism in patients with trapezius myalgia: microdialysis in rest and during exercise. Pain. 2004;112(3):324–334.
    1. Larsson SE, Bodegard L, Henriksson KG, Oberg PA. Chronic trapezius myalgia: morphology and blood flow studied in 17 patients. Acta Orthopaedica Scandinavica. 1990;61(5):394–398.
    1. Kadi F, Waling K, Ahlgren C, et al. Pathological mechanisms implicated in localized female trapezius myalgia. Pain. 1998;78(3):191–196.
    1. Mesquita-Ferrari RA, Martins MD, Silva JA, Jr., et al. Effects of low-level laser therapy on expression of TNF-α and TGF-β in skeletal muscle during the repair process. Lasers in Medical Science. 2011;26(3):335–340.
    1. Araújo FA, Rocha MA, Mendes JB, Andrade SP. Atorvastatin inhibits inflammatory angiogenesis in mice through down regulation of VEGF, TNF-α and TGF-β1. Biomedicine and Pharmacotherapy. 2010;64(1):29–34.
    1. Filippin LI, Moreira AJ, Marroni NP, Xavier RM. Nitric oxide and repair of skeletal muscle injury. Nitric Oxide. 2009;21(3-4):157–163.
    1. Tekin L, Akarsu S, Durmuş O, Cakar E, Dinçer U, Kıralp MZ. The effect of dry needling in the treatment of myofascial pain syndrome: a randomized double-blinded placebo-controlled trial. Clinical Rheumatology. In press.

Source: PubMed

3
S'abonner