Opioid receptor-triggered spinal mTORC1 activation contributes to morphine tolerance and hyperalgesia.

نویسندگان

  • Ji-Tian Xu
  • Jian-Yuan Zhao
  • Xiuli Zhao
  • Davinna Ligons
  • Vinod Tiwari
  • Fidelis E Atianjoh
  • Chun-Yi Lee
  • Lingli Liang
  • Weidong Zang
  • Dolores Njoku
  • Srinivasa N Raja
  • Myron Yaster
  • Yuan-Xiang Tao
چکیده

The development of opioid-induced analgesic tolerance and hyperalgesia is a clinical challenge for managing chronic pain. Adaptive changes in protein translation in the nervous system are thought to promote opioid tolerance and hyperalgesia; however, how opioids drive such changes remains elusive. Here, we report that mammalian target of rapamycin (mTOR), which governs most protein translation, was activated in rat spinal dorsal horn neurons after repeated intrathecal morphine injections. Activation was triggered through μ opioid receptor and mediated by intracellular PI3K/Akt. Spinal mTOR inhibition blocked both induction and maintenance of morphine tolerance and hyperalgesia, without affecting basal pain perception or locomotor functions. These effects were attributed to the attenuation of morphine-induced increases in translation initiation activity, nascent protein synthesis, and expression of some known key tolerance-associated proteins, including neuronal NOS (nNOS), in dorsal horn. Moreover, elevating spinal mTOR activity by knocking down the mTOR-negative regulator TSC2 reduced morphine analgesia, produced pain hypersensitivity, and increased spinal nNOS expression. Our findings implicate the μ opioid receptor-triggered PI3K/Akt/mTOR pathway in promoting morphine-induced spinal protein translation changes and associated morphine tolerance and hyperalgesia. These data suggest that mTOR inhibitors could be explored for prevention and/or reduction of opioid tolerance in chronic pain management.

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

منابع مشابه

Intrathecal rapamycin attenuates morphine-induced analgesic tolerance and hyperalgesia in rats with neuropathic pain.

Repeated and long-term administration of opioids is often accompanied by the initiation of opioid-induced analgesic tolerance and hyperalgesia in chronic pain patients. Our previous studies showed that repeated intrathecal morphine injection activated the mammalian target of rapamycin complex 1 (mTORC1) in spinal dorsal horn neurons and that blocking this activation prevented the initiation of ...

متن کامل

Interaction of NMDA and opioid receptors on thermal hyperalgesia and mechanical allodynia in two models of neuropathic pain

The use of multiple loose ligations of the rat sciatic nerve has been proposed as a model for the study of allodynia and hyperalgesia. This pain hypersensitivity results from both an increase in the peripheral and central sensitization. The evidence indicating that the development of neuropathic thermal hyperalgesia and mechanical allodynia requires activation of spinal cord NMDA receptors. NMD...

متن کامل

Interaction of NMDA and opioid receptors on thermal hyperalgesia and mechanical allodynia in two models of neuropathic pain

The use of multiple loose ligations of the rat sciatic nerve has been proposed as a model for the study of allodynia and hyperalgesia. This pain hypersensitivity results from both an increase in the peripheral and central sensitization. The evidence indicating that the development of neuropathic thermal hyperalgesia and mechanical allodynia requires activation of spinal cord NMDA receptors. NMD...

متن کامل

Complement factor C5a and C5a receptor contribute to morphine tolerance and withdrawal-induced hyperalgesia in rats

Morphine is a potent opioid analgesic. However, the repeated use of morphine causes tolerance and hyperalgesia. Neuroinflammation has been reported to be involved in morphine tolerance and withdrawal-induced hyperalgesia. The complement system is a crucial effector mechanism of immune responses. The present study investigated the roles of complement factor C5a and C5a receptor (C5aR) in the dev...

متن کامل

Epigenetic regulation of spinal cord gene expression contributes to enhanced postoperative pain and analgesic tolerance subsequent to continuous opioid exposure

BACKGROUND Opioids have become the mainstay for treatment of moderate to severe pain and are commonly used to treat surgical pain. While opioid administration has been shown to cause opioid-induced hyperalgesia and tolerance, interactions between opioid administration and surgery with respect to these problematic adaptations have scarcely been addressed. Accumulating evidence suggests opioids a...

متن کامل

ذخیره در منابع من


  با ذخیره ی این منبع در منابع من، دسترسی به آن را برای استفاده های بعدی آسان تر کنید

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

عنوان ژورنال:
  • The Journal of clinical investigation

دوره 124 2  شماره 

صفحات  -

تاریخ انتشار 2014