Skip to main content
Top
Published in: Molecular Pain 1/2008

Open Access 01-12-2008 | Research

Influence of TRPV1 on diabetes-induced alterations in thermal pain sensitivity

Authors: Reddy M Pabbidi, Shuang-Quan Yu, Siying Peng, Romesh Khardori, Mary E Pauza, Louis S Premkumar

Published in: Molecular Pain | Issue 1/2008

Login to get access

Abstract

A common complication associated with diabetes is painful or painless diabetic peripheral neuropathy (DPN). The mechanisms and determinants responsible for these peripheral neuropathies are poorly understood. Using both streptozotocin (STZ)-induced and transgene-mediated murine models of type 1 diabetes (T1D), we demonstrate that Transient Receptor Potential Vanilloid 1 (TRPV1) expression varies with the neuropathic phenotype. We have found that both STZ- and transgene-mediated T1D are associated with two distinct phases of thermal pain sensitivity that parallel changes in TRPV1 as determined by paw withdrawal latency (PWL). An early phase of hyperalgesia and a late phase of hypoalgesia are evident. TRPV1-mediated whole cell currents are larger and smaller in dorsal root ganglion (DRG) neurons collected from hyperalgesic and hypoalgesic mice. Resiniferatoxin (RTX) binding, a measure of TRPV1 expression is increased and decreased in DRG and paw skin of hyperalgesic and hypoalgesic mice, respectively. Immunohistochemical labeling of spinal cord lamina I and II, dorsal root ganglion (DRG), and paw skin from hyperalgesic and hypoalgesic mice reveal increased and decreased TRPV1 expression, respectively. A role for TRPV1 in thermal DPN is further suggested by the failure of STZ treatment to influence thermal nociception in TRPV1 deficient mice. These findings demonstrate that altered TRPV1 expression and function contribute to diabetes-induced changes in thermal perception.
Appendix
Available only for authorised users
Literature
1.
go back to reference Vinik AI, Park TS, Stansberry KB: Diabetic neuropathies. Diabetologia 2000, 43: 957–973. 10.1007/s001250051477CrossRefPubMed Vinik AI, Park TS, Stansberry KB: Diabetic neuropathies. Diabetologia 2000, 43: 957–973. 10.1007/s001250051477CrossRefPubMed
2.
go back to reference Anand P, Terenghi G, Warner G, Kopelman P, Williams-Chestnut RE, Sinicropi DV: The role of endogenous nerve growth factor in human diabetic neuropathy. Nat Med 1996, 2: 703–707. 10.1038/nm0696-703CrossRefPubMed Anand P, Terenghi G, Warner G, Kopelman P, Williams-Chestnut RE, Sinicropi DV: The role of endogenous nerve growth factor in human diabetic neuropathy. Nat Med 1996, 2: 703–707. 10.1038/nm0696-703CrossRefPubMed
3.
go back to reference Sugimoto K, Murakawa Y, Sima AA: Diabetic neuropathy a continuing enigma. Diabetes/Metabolism Res Rev 2000, 16: 408–433. Publisher Full Text 10.1002/1520-7560(200011/12)16:6%3C;408::AID-DMRR158%3E;3.0.CO;2-RCrossRef Sugimoto K, Murakawa Y, Sima AA: Diabetic neuropathy a continuing enigma. Diabetes/Metabolism Res Rev 2000, 16: 408–433. Publisher Full Text 10.1002/1520-7560(200011/12)16:6%3C;408::AID-DMRR158%3E;3.0.CO;2-RCrossRef
4.
go back to reference The diabetes control and complications trial research group: The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med 1993, 329: 977–986. 10.1056/NEJM199309303291401CrossRef The diabetes control and complications trial research group: The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med 1993, 329: 977–986. 10.1056/NEJM199309303291401CrossRef
5.
go back to reference Smith AG, Ramachandran P, Tripp S, Singleton ZR: Epidermal nerve innervation in impaired glucose tolerance and diabetes-associated neuropathy. Neurology 2001, 57: 1701–1704.CrossRefPubMed Smith AG, Ramachandran P, Tripp S, Singleton ZR: Epidermal nerve innervation in impaired glucose tolerance and diabetes-associated neuropathy. Neurology 2001, 57: 1701–1704.CrossRefPubMed
6.
go back to reference Gabra BH, Berthiaume N, Sirois P, Nantel F, Battistini B: The kinin system mediates hyperalgesia through the inducible bradykinin B1 receptor subtype: evidence in various experimental animal models of type 1 and type 2 diabetic neuropathy. Biol Chem 2006, 387: 127–143. 10.1515/BC.2006.018CrossRefPubMed Gabra BH, Berthiaume N, Sirois P, Nantel F, Battistini B: The kinin system mediates hyperalgesia through the inducible bradykinin B1 receptor subtype: evidence in various experimental animal models of type 1 and type 2 diabetic neuropathy. Biol Chem 2006, 387: 127–143. 10.1515/BC.2006.018CrossRefPubMed
7.
go back to reference Pierson CR, Zhang W, Murakawa Y, Sima AAF: Insulin deficiency rather than hyperglycemia accounts for impaired neurotrophic responses and nerve fiber regeneration in type 1 diabetic neuropathy. J Neuropathol Exp Neurol 2003, 62: 260–271.PubMed Pierson CR, Zhang W, Murakawa Y, Sima AAF: Insulin deficiency rather than hyperglycemia accounts for impaired neurotrophic responses and nerve fiber regeneration in type 1 diabetic neuropathy. J Neuropathol Exp Neurol 2003, 62: 260–271.PubMed
8.
go back to reference Sima AAF: Diabetes underlies common neurological disorders. Ann Neurol 2004, 56: 459–461. 10.1002/ana.20249CrossRefPubMed Sima AAF: Diabetes underlies common neurological disorders. Ann Neurol 2004, 56: 459–461. 10.1002/ana.20249CrossRefPubMed
9.
go back to reference Brussee V, Cunningham FA, Zochodne DW: Direct insulin signaling of neurons reverses diabetic neuropathy. Diabetes 2004, 53: 1824–1830. 10.2337/diabetes.53.7.1824CrossRefPubMed Brussee V, Cunningham FA, Zochodne DW: Direct insulin signaling of neurons reverses diabetic neuropathy. Diabetes 2004, 53: 1824–1830. 10.2337/diabetes.53.7.1824CrossRefPubMed
10.
11.
12.
go back to reference Julius D, Basbaum AI: Acid potentiation of the capsaicin receptor determined by a key extracellular site. Proc Natl Acad Sci USA 2001, 97: 8134–8139. Julius D, Basbaum AI: Acid potentiation of the capsaicin receptor determined by a key extracellular site. Proc Natl Acad Sci USA 2001, 97: 8134–8139.
13.
go back to reference Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D: The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 1997, 389: 816–824. 10.1038/39807CrossRefPubMed Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D: The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 1997, 389: 816–824. 10.1038/39807CrossRefPubMed
14.
go back to reference Chuang HH, Prescott ED, Kong H, Shields S, Jordt SE, Basbaum AI, Chao MV, Julius D: Bradykinin and nerve growth factor release the capsaicin receptor from Ptdlns (4,5) P2-Mediated inhibition. Nature 2001, 411: 957–962. 10.1038/35082088CrossRefPubMed Chuang HH, Prescott ED, Kong H, Shields S, Jordt SE, Basbaum AI, Chao MV, Julius D: Bradykinin and nerve growth factor release the capsaicin receptor from Ptdlns (4,5) P2-Mediated inhibition. Nature 2001, 411: 957–962. 10.1038/35082088CrossRefPubMed
15.
go back to reference Huang SM, Bisogno T, Trevisani M, Al-Hayani A, De Petrocellis L, Fezza F, Tognetto M, Petros TJ, Krey JF, Chu CJ, Miller JD, Davies SN, Geppetti P, Walker JM, Di Marzo V: An endogenous capsaicin-like substance with high potency at recombinant and native vanilloid VR1 receptors. Proc Natl Acad Sci USA 2002, 99: 8400–8405. 10.1073/pnas.122196999PubMedCentralCrossRefPubMed Huang SM, Bisogno T, Trevisani M, Al-Hayani A, De Petrocellis L, Fezza F, Tognetto M, Petros TJ, Krey JF, Chu CJ, Miller JD, Davies SN, Geppetti P, Walker JM, Di Marzo V: An endogenous capsaicin-like substance with high potency at recombinant and native vanilloid VR1 receptors. Proc Natl Acad Sci USA 2002, 99: 8400–8405. 10.1073/pnas.122196999PubMedCentralCrossRefPubMed
16.
go back to reference Van der Stelt M, Di Marzo V: Putative endogenous ligands of transient receptor potential vanilloid 1 channels. Eur J Biochem 2004, 271: 1827–1834. 10.1111/j.1432-1033.2004.04081.xCrossRefPubMed Van der Stelt M, Di Marzo V: Putative endogenous ligands of transient receptor potential vanilloid 1 channels. Eur J Biochem 2004, 271: 1827–1834. 10.1111/j.1432-1033.2004.04081.xCrossRefPubMed
17.
go back to reference Lopshire JC, Nicol GD: The cAMP transduction cascade mediates the prostaglandin E2 enhancement of the capsaicin-elicited current in rat sensory neurons, whole-cell and single-channel studies. J Neurosci 1998, 18: 6081–6092.PubMed Lopshire JC, Nicol GD: The cAMP transduction cascade mediates the prostaglandin E2 enhancement of the capsaicin-elicited current in rat sensory neurons, whole-cell and single-channel studies. J Neurosci 1998, 18: 6081–6092.PubMed
18.
go back to reference Moriyama T, Higashim T, Togashi K, Iida T, Segi E, Sugimoto Y, Tominaga T, Narumiya S, Tominaga M: Sensitization of TRPV1 by EP1and IP reveals peripheral nociceptive mechanism of prostaglandins. Mol Pain 2005, 1: 3. 10.1186/1744-8069-1-3PubMedCentralCrossRefPubMed Moriyama T, Higashim T, Togashi K, Iida T, Segi E, Sugimoto Y, Tominaga T, Narumiya S, Tominaga M: Sensitization of TRPV1 by EP1and IP reveals peripheral nociceptive mechanism of prostaglandins. Mol Pain 2005, 1: 3. 10.1186/1744-8069-1-3PubMedCentralCrossRefPubMed
19.
go back to reference Cesare P, McNaughton P: A novel heat-activated current in nociceptive neurons and its sensitization by bradykinin. Proc Natl Acad Sci USA 1996, 93: 15435–15439. 10.1073/pnas.93.26.15435PubMedCentralCrossRefPubMed Cesare P, McNaughton P: A novel heat-activated current in nociceptive neurons and its sensitization by bradykinin. Proc Natl Acad Sci USA 1996, 93: 15435–15439. 10.1073/pnas.93.26.15435PubMedCentralCrossRefPubMed
20.
go back to reference Premkumar LS, Ahern GP: Induction of vanilloid receptor channel activity by protein kinase C. Nature 2000, 408: 985–990. 10.1038/35050121CrossRefPubMed Premkumar LS, Ahern GP: Induction of vanilloid receptor channel activity by protein kinase C. Nature 2000, 408: 985–990. 10.1038/35050121CrossRefPubMed
21.
go back to reference Sugiura T, Tominaga M, Katsuya H, Mizumura K: Bradykinin lowers the threshold temperature for heat activation of vanilloid receptor 1. J Neurophysiol 2002, 88: 544–548.PubMed Sugiura T, Tominaga M, Katsuya H, Mizumura K: Bradykinin lowers the threshold temperature for heat activation of vanilloid receptor 1. J Neurophysiol 2002, 88: 544–548.PubMed
22.
go back to reference Hu HJ, Bhave G, Gereau RWT: Prostaglandin and protein kinase A-dependent modulation of vanilloid receptor function by metabotropic glutamate receptor 5: potential mechanism for thermal hyperalgesia. J Neurosci 2002, 22: 7444–7452.PubMed Hu HJ, Bhave G, Gereau RWT: Prostaglandin and protein kinase A-dependent modulation of vanilloid receptor function by metabotropic glutamate receptor 5: potential mechanism for thermal hyperalgesia. J Neurosci 2002, 22: 7444–7452.PubMed
23.
go back to reference Sugiura T, Bielefeldt K, Gebhart GF: TRPV1 function in mouse colon sensory neurons is enhanced by metabotropic 5-hydroxytryptamine receptor activation. J Neurosci 2004, 24: 9521–9530. 10.1523/JNEUROSCI.2639-04.2004CrossRef Sugiura T, Bielefeldt K, Gebhart GF: TRPV1 function in mouse colon sensory neurons is enhanced by metabotropic 5-hydroxytryptamine receptor activation. J Neurosci 2004, 24: 9521–9530. 10.1523/JNEUROSCI.2639-04.2004CrossRef
24.
go back to reference Kim BM, Lee SH, Shim WS, Oh U: Histamine-induced Ca2+influx via the PLA(2)/lipoxygenase/TRPV1 pathway in rat sensory neurons. Neurosci Lett 2004, 361: 159–162. 10.1016/j.neulet.2004.01.019CrossRefPubMed Kim BM, Lee SH, Shim WS, Oh U: Histamine-induced Ca2+influx via the PLA(2)/lipoxygenase/TRPV1 pathway in rat sensory neurons. Neurosci Lett 2004, 361: 159–162. 10.1016/j.neulet.2004.01.019CrossRefPubMed
25.
go back to reference Kress M, Guenther S: Role of [Ca2+]iin the ATP-induced heat sensitization process of rat nociceptive neurons. J Neurophysiol 1999, 81: 2612–2619.PubMed Kress M, Guenther S: Role of [Ca2+]iin the ATP-induced heat sensitization process of rat nociceptive neurons. J Neurophysiol 1999, 81: 2612–2619.PubMed
26.
go back to reference Tominaga M, Wada M, Masu M: Potentiation of capsaicin receptor activity by metabotropic ATP receptors as a possible mechanism for ATP-evoked pain and hyperalgesia. Proc Natl Acad Sci USA 2001, 98: 6951–6956. 10.1073/pnas.111025298PubMedCentralCrossRefPubMed Tominaga M, Wada M, Masu M: Potentiation of capsaicin receptor activity by metabotropic ATP receptors as a possible mechanism for ATP-evoked pain and hyperalgesia. Proc Natl Acad Sci USA 2001, 98: 6951–6956. 10.1073/pnas.111025298PubMedCentralCrossRefPubMed
27.
go back to reference Kwak J, Wang MH, Hwang SW, Kim TY, Lee SY, Oh U: Intracellular ATP increases capsaicin-activated channel activity by interacting with nucleotide-binding domains. J Neurosci 2000, 20: 8298–8304.PubMed Kwak J, Wang MH, Hwang SW, Kim TY, Lee SY, Oh U: Intracellular ATP increases capsaicin-activated channel activity by interacting with nucleotide-binding domains. J Neurosci 2000, 20: 8298–8304.PubMed
28.
go back to reference Amadesi S, Nie J, Vergnolle N, Cottrell GS, Grady EF, Trevisani M, Manni C, Geppetti P, McRoberts JA, Ennes H, Davis JB, Mayer EA, Bunnett NWJ: Protease-Activated Receptor 2 sensitizes the capsaicin receptor transient receptor potential vanilloid receptor 1 to induce hyperalgesia. J Neurosci 2004, 24: 4300–4312. 10.1523/JNEUROSCI.5679-03.2004CrossRefPubMed Amadesi S, Nie J, Vergnolle N, Cottrell GS, Grady EF, Trevisani M, Manni C, Geppetti P, McRoberts JA, Ennes H, Davis JB, Mayer EA, Bunnett NWJ: Protease-Activated Receptor 2 sensitizes the capsaicin receptor transient receptor potential vanilloid receptor 1 to induce hyperalgesia. J Neurosci 2004, 24: 4300–4312. 10.1523/JNEUROSCI.5679-03.2004CrossRefPubMed
29.
go back to reference Dai Y, Moriyama T, Higashi T, Togashi K, Kobayashi K, Yamanaka H, Tominaga M, Noguchi K: Proteinase-activated receptor 2-mediated potentiation of transient receptor potential vanilloid subfamily 1 activity reveals a mechanism for proteinase-induced inflammatory pain. J Neurosci 2004, 24: 4293–4299. 10.1523/JNEUROSCI.0454-04.2004CrossRefPubMed Dai Y, Moriyama T, Higashi T, Togashi K, Kobayashi K, Yamanaka H, Tominaga M, Noguchi K: Proteinase-activated receptor 2-mediated potentiation of transient receptor potential vanilloid subfamily 1 activity reveals a mechanism for proteinase-induced inflammatory pain. J Neurosci 2004, 24: 4293–4299. 10.1523/JNEUROSCI.0454-04.2004CrossRefPubMed
30.
go back to reference Shu Q, Mendell LM: Acute sensitization by NGF of the response of small-diameter sensory neurons to capsaicin. J Neurophysiol 2001, 86: 2931–2938.PubMed Shu Q, Mendell LM: Acute sensitization by NGF of the response of small-diameter sensory neurons to capsaicin. J Neurophysiol 2001, 86: 2931–2938.PubMed
31.
go back to reference Capsaicin Study Group: Treatment of painful diabetic neuropathy with topical capsaicin: a multicenter, double-blind, vehicle-controlled study. Arch Intern Med 1991, 151: 2225–2229. 10.1001/archinte.151.11.2225CrossRef Capsaicin Study Group: Treatment of painful diabetic neuropathy with topical capsaicin: a multicenter, double-blind, vehicle-controlled study. Arch Intern Med 1991, 151: 2225–2229. 10.1001/archinte.151.11.2225CrossRef
32.
go back to reference Forst T, Pohlmann T, Kunt T, Goitom K, Schulz G, Lobig M, Engelbach M, Beyer J, Pfutzner A: The influence of local capsaicin treatment on small nerve fiber function and neurovascular control in symptomatic diabetic neuropathy. Acta Diabetologica 2002, 39: 1–6. 10.1007/s005920200005CrossRefPubMed Forst T, Pohlmann T, Kunt T, Goitom K, Schulz G, Lobig M, Engelbach M, Beyer J, Pfutzner A: The influence of local capsaicin treatment on small nerve fiber function and neurovascular control in symptomatic diabetic neuropathy. Acta Diabetologica 2002, 39: 1–6. 10.1007/s005920200005CrossRefPubMed
33.
go back to reference Kamei J, Zushida K, Mortia K, Sasaki M, Tanaka S: Role of vanilloid VR1 receptor in thermal allodynia and hyperalgesia in diabetic mice. Eur J Pharmacol 2001, 422: 83–86. 10.1016/S0014-2999(01)01059-7CrossRefPubMed Kamei J, Zushida K, Mortia K, Sasaki M, Tanaka S: Role of vanilloid VR1 receptor in thermal allodynia and hyperalgesia in diabetic mice. Eur J Pharmacol 2001, 422: 83–86. 10.1016/S0014-2999(01)01059-7CrossRefPubMed
34.
go back to reference Rashid MH, Ueda H: Nonopioid and neuropathy-specific analgesic action of the nootropic drug nefiracetam in mice. J Pharmacol Exp Ther 2002, 303: 226–231. 10.1124/jpet.102.037952CrossRefPubMed Rashid MH, Ueda H: Nonopioid and neuropathy-specific analgesic action of the nootropic drug nefiracetam in mice. J Pharmacol Exp Ther 2002, 303: 226–231. 10.1124/jpet.102.037952CrossRefPubMed
35.
go back to reference Kirberg J, Berns A, von Boehmer H: Peripheral T Cell survival requires continual ligation of the T cell receptor to major histocompatibility complex-encoded molecules. J Exp Med 1997, 186: 1269–1275. 10.1084/jem.186.8.1269PubMedCentralCrossRefPubMed Kirberg J, Berns A, von Boehmer H: Peripheral T Cell survival requires continual ligation of the T cell receptor to major histocompatibility complex-encoded molecules. J Exp Med 1997, 186: 1269–1275. 10.1084/jem.186.8.1269PubMedCentralCrossRefPubMed
36.
go back to reference Lo D, Freedman J, Hesse S, Palmiter RD, Brinster RL, Sherman LA: Peripheral tolerance to an islet cell-specific hemagglutinin transgene affects both CD4+and CD8+T cells. Eur J Immunol 1992, 22: 1013–1032. 10.1002/eji.1830220421CrossRefPubMed Lo D, Freedman J, Hesse S, Palmiter RD, Brinster RL, Sherman LA: Peripheral tolerance to an islet cell-specific hemagglutinin transgene affects both CD4+and CD8+T cells. Eur J Immunol 1992, 22: 1013–1032. 10.1002/eji.1830220421CrossRefPubMed
37.
go back to reference Smith SS, Patterson T, Pauza ME: Transgenic Ly-49A inhibits antigen-driven T cell activation and delays diabetes. J Immunol 2005, 174: 3897–3905.CrossRefPubMed Smith SS, Patterson T, Pauza ME: Transgenic Ly-49A inhibits antigen-driven T cell activation and delays diabetes. J Immunol 2005, 174: 3897–3905.CrossRefPubMed
38.
go back to reference Caterina MJ, Leffler A, Malmberg AB, Martin WJ, Trafton J, Petersen-Zeitz KR, Koltzenburg M, Basbaum AI, Julius D: Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science 2000, 288: 306–313. 10.1126/science.288.5464.306CrossRefPubMed Caterina MJ, Leffler A, Malmberg AB, Martin WJ, Trafton J, Petersen-Zeitz KR, Koltzenburg M, Basbaum AI, Julius D: Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science 2000, 288: 306–313. 10.1126/science.288.5464.306CrossRefPubMed
39.
go back to reference 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: Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia. Nature 2000, 405: 183–187. 10.1038/35012076CrossRefPubMed 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: Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia. Nature 2000, 405: 183–187. 10.1038/35012076CrossRefPubMed
40.
go back to reference Kamei U, Ohhashi Y, Aoki T, Kasuya Y: Streptozotocin-induced diabetes in mice reduces the nociceptive threshold, as recognized after application of noxious mechanical stimuli but not of thermal stimuli. Pharmacol Biochem Behav 1991, 39: 541–544. 10.1016/0091-3057(91)90224-PCrossRefPubMed Kamei U, Ohhashi Y, Aoki T, Kasuya Y: Streptozotocin-induced diabetes in mice reduces the nociceptive threshold, as recognized after application of noxious mechanical stimuli but not of thermal stimuli. Pharmacol Biochem Behav 1991, 39: 541–544. 10.1016/0091-3057(91)90224-PCrossRefPubMed
41.
go back to reference Hong S, Wiley JW: Early painful diabetic neuropathy is associated with differential changes in the expression and function of vanilloid receptor 1. J Biol Chem 2005, 280: 618–627.CrossRefPubMed Hong S, Wiley JW: Early painful diabetic neuropathy is associated with differential changes in the expression and function of vanilloid receptor 1. J Biol Chem 2005, 280: 618–627.CrossRefPubMed
42.
go back to reference Puntambekar P, Van Buren J, Raisinghani M, Premkumar LS, Ramkumar V: Direct interaction of adenosine with the TRPV1 channel protein. J Neurosci 2004, 24: 3663–3671. 10.1523/JNEUROSCI.4773-03.2004CrossRefPubMed Puntambekar P, Van Buren J, Raisinghani M, Premkumar LS, Ramkumar V: Direct interaction of adenosine with the TRPV1 channel protein. J Neurosci 2004, 24: 3663–3671. 10.1523/JNEUROSCI.4773-03.2004CrossRefPubMed
43.
go back to reference Szallasi A, Blumberg PM, Annicelli LL, Krause JE, Cortright DN: The cloned rat vanilloid receptor VR1 mediates both R-type binding and C-type calcium response in dorsal root ganglion neurons. Mol Pharmacol 1999, 56: 581–587.PubMed Szallasi A, Blumberg PM, Annicelli LL, Krause JE, Cortright DN: The cloned rat vanilloid receptor VR1 mediates both R-type binding and C-type calcium response in dorsal root ganglion neurons. Mol Pharmacol 1999, 56: 581–587.PubMed
44.
go back to reference Rashid MH, Inoue M, Kondo S, Kawashima T, Bakoshi S, Ueda H: Novel expression of vanilloid receptor 1 on capsaicin-insensitive fibers accounts for the analgesic effect of capsaicin cream in neuropathic pain. J Pharmacol Exp Ther 2003, 304: 940–948. 10.1124/jpet.102.046250CrossRefPubMed Rashid MH, Inoue M, Kondo S, Kawashima T, Bakoshi S, Ueda H: Novel expression of vanilloid receptor 1 on capsaicin-insensitive fibers accounts for the analgesic effect of capsaicin cream in neuropathic pain. J Pharmacol Exp Ther 2003, 304: 940–948. 10.1124/jpet.102.046250CrossRefPubMed
45.
go back to reference Kishi M, Tanabe J, Schmelzer JD, Low PA: Morphometry of dorsal root ganglion in chronic experimental diabetic neuropathy. Diabetes 2002, 51: 819–824. 10.2337/diabetes.51.3.819CrossRefPubMed Kishi M, Tanabe J, Schmelzer JD, Low PA: Morphometry of dorsal root ganglion in chronic experimental diabetic neuropathy. Diabetes 2002, 51: 819–824. 10.2337/diabetes.51.3.819CrossRefPubMed
46.
go back to reference Sullivan KA, Feldman EL: New developments in diabetic neuropathy. Curr Opin Neurol 2005, 18: 586–590. 10.1097/01.wco.0000178825.56414.52CrossRefPubMed Sullivan KA, Feldman EL: New developments in diabetic neuropathy. Curr Opin Neurol 2005, 18: 586–590. 10.1097/01.wco.0000178825.56414.52CrossRefPubMed
47.
go back to reference Pauza ME, Nguyen A, Wolfe T, Ho IC, Glimcher LH, Herrath MV, Lo D: Variable effects of transgenic c-maf on autoimmune diabetes. Diabetes 2001, 50: 39–46. 10.2337/diabetes.50.1.39CrossRefPubMed Pauza ME, Nguyen A, Wolfe T, Ho IC, Glimcher LH, Herrath MV, Lo D: Variable effects of transgenic c-maf on autoimmune diabetes. Diabetes 2001, 50: 39–46. 10.2337/diabetes.50.1.39CrossRefPubMed
48.
go back to reference Kolta MG, Ngong JM, Rutledge LP, Pierzchala K, VanLoon GR: Endogenous opioid peptide mediation of hypoalgesic response in long-term diabetic rats. Neuropeptides 1996, 30: 335–344. 10.1016/S0143-4179(96)90022-0CrossRefPubMed Kolta MG, Ngong JM, Rutledge LP, Pierzchala K, VanLoon GR: Endogenous opioid peptide mediation of hypoalgesic response in long-term diabetic rats. Neuropeptides 1996, 30: 335–344. 10.1016/S0143-4179(96)90022-0CrossRefPubMed
49.
go back to reference Calcutt NA, Freshwater JD, Mizisin AP: Prevention of sensory disorders in diabetic Sprague-Dawley rats by aldose reductase inhibition or treatment with ciliary neurotrophic factor. Diabetologia 2004, 47: 718–724. 10.1007/s00125-004-1354-2CrossRefPubMed Calcutt NA, Freshwater JD, Mizisin AP: Prevention of sensory disorders in diabetic Sprague-Dawley rats by aldose reductase inhibition or treatment with ciliary neurotrophic factor. Diabetologia 2004, 47: 718–724. 10.1007/s00125-004-1354-2CrossRefPubMed
50.
go back to reference Van Buren JJ, Bhat S, Rotello R, Pauza ME, Premkumar LS: Sensitization and translocation of TRPV1 by insulin and IGF-I. Mol Pain 2005, 1: 1–17. 10.1186/1744-8069-1-17CrossRef Van Buren JJ, Bhat S, Rotello R, Pauza ME, Premkumar LS: Sensitization and translocation of TRPV1 by insulin and IGF-I. Mol Pain 2005, 1: 1–17. 10.1186/1744-8069-1-17CrossRef
51.
go back to reference Schmeichel AM, Schmelzer JD, Low PA: Oxidative injury and apoptosis of dorsal root ganglion neurons in chronic experimental diabetic neuropathy. Diabetes 2003, 52: 165–171. 10.2337/diabetes.52.1.165CrossRefPubMed Schmeichel AM, Schmelzer JD, Low PA: Oxidative injury and apoptosis of dorsal root ganglion neurons in chronic experimental diabetic neuropathy. Diabetes 2003, 52: 165–171. 10.2337/diabetes.52.1.165CrossRefPubMed
52.
go back to reference Suzukawa K, Miura K, Mitsushita J, Resau J, Hirose K, Crystal R, Kamata T: Nerve growth factor-induced neuronal differentiation requires generation of Rac1-regulated reactive oxygen species. J Biol Chem 2000, 275: 175–178. 10.1074/jbc.275.18.13175CrossRef Suzukawa K, Miura K, Mitsushita J, Resau J, Hirose K, Crystal R, Kamata T: Nerve growth factor-induced neuronal differentiation requires generation of Rac1-regulated reactive oxygen species. J Biol Chem 2000, 275: 175–178. 10.1074/jbc.275.18.13175CrossRef
53.
go back to reference Pabbidi RM, Cao DS, Parihar A, Pauza ME, Premkumar LS: Direct role of streptozotocin in inducing thermal hyperalgesia by enhanced expression of TRPV1 in sensory neurons. Mol Pharmacol 2008, in press. Pabbidi RM, Cao DS, Parihar A, Pauza ME, Premkumar LS: Direct role of streptozotocin in inducing thermal hyperalgesia by enhanced expression of TRPV1 in sensory neurons. Mol Pharmacol 2008, in press.
54.
go back to reference Di Marzo V, Blumberg PM, Szallasi A: Endovanilloid signaling in pain. Curr Opin Neurobiol 2002, 12: 372–379. 10.1016/S0959-4388(02)00340-9CrossRefPubMed Di Marzo V, Blumberg PM, Szallasi A: Endovanilloid signaling in pain. Curr Opin Neurobiol 2002, 12: 372–379. 10.1016/S0959-4388(02)00340-9CrossRefPubMed
55.
go back to reference Ji R, Samad TA, Jin S, Schomll R, Woolf CJ: p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 2002, 36: 57–68. 10.1016/S0896-6273(02)00908-XCrossRefPubMed Ji R, Samad TA, Jin S, Schomll R, Woolf CJ: p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 2002, 36: 57–68. 10.1016/S0896-6273(02)00908-XCrossRefPubMed
56.
go back to reference Eicheberg J: Protein kinase C changes in diabetes: is the concept relevant to neuropathy? Int Rev Neurobiol 2002, 50: 61–82.CrossRef Eicheberg J: Protein kinase C changes in diabetes: is the concept relevant to neuropathy? Int Rev Neurobiol 2002, 50: 61–82.CrossRef
57.
go back to reference Way KJ, Katai N, King GL: Protein kinase C and the development of diabetic vascular complications. Diabet Med 2001, 18: 945–959. 10.1046/j.0742-3071.2001.00638.xCrossRefPubMed Way KJ, Katai N, King GL: Protein kinase C and the development of diabetic vascular complications. Diabet Med 2001, 18: 945–959. 10.1046/j.0742-3071.2001.00638.xCrossRefPubMed
58.
go back to reference Mezey E, Toth ZE, Cortright DN, Arzubi MK, Krause JE, Elde R, Guo A, Blumberg PM, Szallasi A: Distribution of mRNA for vanilloid receptor subtype 1 (VR1), and VR1-like immunoreactivity, in the central nervous system of the rat and human. Proc Natl Acad Sci USA 2000, 97: 3655–3660. 10.1073/pnas.060496197PubMedCentralCrossRefPubMed Mezey E, Toth ZE, Cortright DN, Arzubi MK, Krause JE, Elde R, Guo A, Blumberg PM, Szallasi A: Distribution of mRNA for vanilloid receptor subtype 1 (VR1), and VR1-like immunoreactivity, in the central nervous system of the rat and human. Proc Natl Acad Sci USA 2000, 97: 3655–3660. 10.1073/pnas.060496197PubMedCentralCrossRefPubMed
59.
go back to reference Sikand P, Premkumar LS: Potentiation of Glutamatergic Synaptic Transmission by Protein Kinase C Mediated Sensitization of TRPV1 at the First Sensory Synapse. J Physiol 2007, 2: 631–647. 10.1113/jphysiol.2006.118620CrossRef Sikand P, Premkumar LS: Potentiation of Glutamatergic Synaptic Transmission by Protein Kinase C Mediated Sensitization of TRPV1 at the First Sensory Synapse. J Physiol 2007, 2: 631–647. 10.1113/jphysiol.2006.118620CrossRef
60.
go back to reference Marinelli S, Di Marzo V, Berretta N, Matias I, Maccarrone M, Bernardi G, Mercuri NB: Presynaptic facilitation of glutamatergic synapses to dopaminergic neurons of the rat substantia nigra by endogenous stimulation of vanilloid receptors. J Neurosci 2003, 23: 3136–3144.PubMed Marinelli S, Di Marzo V, Berretta N, Matias I, Maccarrone M, Bernardi G, Mercuri NB: Presynaptic facilitation of glutamatergic synapses to dopaminergic neurons of the rat substantia nigra by endogenous stimulation of vanilloid receptors. J Neurosci 2003, 23: 3136–3144.PubMed
61.
go back to reference Tognetto M, Amadesi S, Harrison S, Creminon C, Trevisani M, Carreras M, Matera M, Geppetti P, Bianchi A: Anandamide excites central terminals of dorsal root ganglion neurons via vanilloid receptor-1 activation. J Neurosci 2001, 21: 1104–1109.PubMed Tognetto M, Amadesi S, Harrison S, Creminon C, Trevisani M, Carreras M, Matera M, Geppetti P, Bianchi A: Anandamide excites central terminals of dorsal root ganglion neurons via vanilloid receptor-1 activation. J Neurosci 2001, 21: 1104–1109.PubMed
62.
go back to reference Razavi R, Chan Y, Afifiyan FN, Liu XJ, Wan X, Yantha J, Tsui H, Tang L, Tsai S, Santamaria P, Driver JP, Serreze D, Salter MW, Dosch HD: TRPV1+Sensory Neurons Control β Cell Stress and Islet Inflammation in Autoimmune Diabetes. Cell 2006, 127: 1123–1135. 10.1016/j.cell.2006.10.038CrossRefPubMed Razavi R, Chan Y, Afifiyan FN, Liu XJ, Wan X, Yantha J, Tsui H, Tang L, Tsai S, Santamaria P, Driver JP, Serreze D, Salter MW, Dosch HD: TRPV1+Sensory Neurons Control β Cell Stress and Islet Inflammation in Autoimmune Diabetes. Cell 2006, 127: 1123–1135. 10.1016/j.cell.2006.10.038CrossRefPubMed
63.
go back to reference Birder LA, Kanai AJ, De Groat WC, Kiss S, Nealen ML, Burke NE, Dineley KE, Watkins S, Reynolds IJ, Caterina MJ: Vanilloid receptor expression suggests a sensory role for urinary bladder epithelial cells. Proc Natl Acad Sci USA 2001, 98: 13396–13401. 10.1073/pnas.231243698PubMedCentralCrossRefPubMed Birder LA, Kanai AJ, De Groat WC, Kiss S, Nealen ML, Burke NE, Dineley KE, Watkins S, Reynolds IJ, Caterina MJ: Vanilloid receptor expression suggests a sensory role for urinary bladder epithelial cells. Proc Natl Acad Sci USA 2001, 98: 13396–13401. 10.1073/pnas.231243698PubMedCentralCrossRefPubMed
64.
go back to reference Birder LA, Nakamura Y, Kiss S, Nealen MI, Kanai AJ, Wang E, Ruiz G, De Groat WC, Apodaca G, Watkins S, Caterina MJ: Altered urinary bladder function in mice lacking the vanilloid receptor TRPV1. Nature 2002, 5: 856–860. Birder LA, Nakamura Y, Kiss S, Nealen MI, Kanai AJ, Wang E, Ruiz G, De Groat WC, Apodaca G, Watkins S, Caterina MJ: Altered urinary bladder function in mice lacking the vanilloid receptor TRPV1. Nature 2002, 5: 856–860.
66.
go back to reference Zygmunt PM, Petersson J, Andersson DA, Chuang H, Sorgard M, Di Marzo V, Julius D, Hogestatt ED: Vanilloid receptors on sensory nerves mediate the vasodilator action of anandamide. Nature 1999, 400: 452–457. 10.1038/22761CrossRefPubMed Zygmunt PM, Petersson J, Andersson DA, Chuang H, Sorgard M, Di Marzo V, Julius D, Hogestatt ED: Vanilloid receptors on sensory nerves mediate the vasodilator action of anandamide. Nature 1999, 400: 452–457. 10.1038/22761CrossRefPubMed
67.
go back to reference Premkumar LS, Raisinghani M: Nociceptors in cardiovascular functions: complex interplay as a result of cyclooxygenase inhibition. Mol Pain 2006, 2: 26. 10.1186/1744-8069-2-26PubMedCentralCrossRefPubMed Premkumar LS, Raisinghani M: Nociceptors in cardiovascular functions: complex interplay as a result of cyclooxygenase inhibition. Mol Pain 2006, 2: 26. 10.1186/1744-8069-2-26PubMedCentralCrossRefPubMed
Metadata
Title
Influence of TRPV1 on diabetes-induced alterations in thermal pain sensitivity
Authors
Reddy M Pabbidi
Shuang-Quan Yu
Siying Peng
Romesh Khardori
Mary E Pauza
Louis S Premkumar
Publication date
01-12-2008
Publisher
BioMed Central
Published in
Molecular Pain / Issue 1/2008
Electronic ISSN: 1744-8069
DOI
https://doi.org/10.1186/1744-8069-4-9

Other articles of this Issue 1/2008

Molecular Pain 1/2008 Go to the issue