Skip to main content
Top
Published in: Journal of Translational Medicine 1/2016

Open Access 01-12-2016 | Research

Hsa-miR-34a mediated repression of corticotrophin releasing hormone receptor 1 regulates pro-opiomelanocortin expression in patients with complex regional pain syndrome

Authors: Botros B. Shenoda, Guillermo M. Alexander, Seena K. Ajit

Published in: Journal of Translational Medicine | Issue 1/2016

Login to get access

Abstract

Background

Ketamine provides relief for a subset of patients with complex regional pain syndrome (CRPS). The poor responders had a lower body mass index (BMI) relative to responders. Regulation of proopiomelanocortin (POMC) expression is crucial in normal body weight homeostasis. The main objectives of this study were to investigate the mechanisms underlying lower BMI characterizing CRPS patients responding poorly to intravenous ketamine therapy and identify potential biomarkers for predicting response.

Methods

We investigated POMC transcript levels in blood from CRPS patients grouped as responders and poor responders to ketamine therapy. Plasma levels of β-endorphin, ACTH and α-MSH were measured by ELISA. We previously identified differential expression of small noncoding microRNA hsa-miR-34a in blood between responders and poor responders. We investigated whether a 11-fold downregulation of hsa-miR-34a in poor responders relative to responders is contributing to the differences in POMC levels by targeting POMC regulator CRHR1. Binding of miR-34a to CRHR1 was assessed using reporter assay; changes in mRNA and protein levels of CRHR1 were used to determine the regulation of CRHR1 by miR-34a. RNA from blood of CRPS and control subjects were used for quantitative PCR for CRHR1.

Results

Though ketamine treatment did not alter POMC expression, poor responders had higher levels of POMC mRNA than responders, both before and after treatment. Corticotropin-releasing hormone (CRH) is a key regulator of POMC expression and the biological effects are mediated through its receptor corticotrophin releasing hormone receptor 1 (CRHR1). We show that hsa-miR-34a is a negative regulator of CRHR1; overexpression of hsa-miR-34a in Jurkat cells resulted in reduction of CRH-mediated POMC expression. Poor responders had higher expression of CRHR1 transcripts than responders, indicating a regulatory role for miR-34a. In addition, we found positive correlations between the pretreatment levels of miR-34a to BMI and response to ketamine therapy.

Conclusions

Our findings indicate a mechanism by which hsa-miR-34a can regulate the CRH/CRHR1/POMC axis and may influence BMI. Studies in larger patient cohorts are required to confirm the biomarker utility of circulating hsa-miR-34a levels in predicting treatment response to ketamine therapy.
Appendix
Available only for authorised users
Literature
1.
go back to reference Borchers AT, Gershwin ME. Complex regional pain syndrome: a comprehensive and critical review. Autoimmun Rev. 2014;13(3):242–65.CrossRefPubMed Borchers AT, Gershwin ME. Complex regional pain syndrome: a comprehensive and critical review. Autoimmun Rev. 2014;13(3):242–65.CrossRefPubMed
2.
go back to reference Gierthmuhlen J, Binder A, Baron R. Mechanism-based treatment in complex regional pain syndromes. Nat Rev Neurol. 2014;10(9):518–28.CrossRefPubMed Gierthmuhlen J, Binder A, Baron R. Mechanism-based treatment in complex regional pain syndromes. Nat Rev Neurol. 2014;10(9):518–28.CrossRefPubMed
3.
go back to reference Birklein F, Schlereth T. Complex regional pain syndrome-significant progress in understanding. Pain. 2015;156(Suppl 1):S94–103.CrossRefPubMed Birklein F, Schlereth T. Complex regional pain syndrome-significant progress in understanding. Pain. 2015;156(Suppl 1):S94–103.CrossRefPubMed
4.
go back to reference Schwartzman RJ, Alexander GM, Grothusen JR. The use of ketamine in complex regional pain syndrome: possible mechanisms. Expert Rev Neurother. 2011;11(5):719–34.CrossRefPubMed Schwartzman RJ, Alexander GM, Grothusen JR. The use of ketamine in complex regional pain syndrome: possible mechanisms. Expert Rev Neurother. 2011;11(5):719–34.CrossRefPubMed
5.
go back to reference Dahan A, et al. Population pharmacokinetic-pharmacodynamic modeling of ketamine-induced pain relief of chronic pain. Eur J Pain. 2011;15(3):258–67.CrossRefPubMed Dahan A, et al. Population pharmacokinetic-pharmacodynamic modeling of ketamine-induced pain relief of chronic pain. Eur J Pain. 2011;15(3):258–67.CrossRefPubMed
6.
go back to reference Douglas SR, et al. Analgesic Response to Intravenous Ketamine Is Linked to a Circulating microRNA Signature in Female Patients With Complex Regional Pain Syndrome. J Pain. 2015;16(9):814–24.CrossRefPubMed Douglas SR, et al. Analgesic Response to Intravenous Ketamine Is Linked to a Circulating microRNA Signature in Female Patients With Complex Regional Pain Syndrome. J Pain. 2015;16(9):814–24.CrossRefPubMed
7.
go back to reference Bicknell AB. The tissue-specific processing of pro-opiomelanocortin. J Neuroendocrinol. 2008;20(6):692–9.CrossRefPubMed Bicknell AB. The tissue-specific processing of pro-opiomelanocortin. J Neuroendocrinol. 2008;20(6):692–9.CrossRefPubMed
8.
9.
go back to reference Smith AI, Funder JW. Proopiomelanocortin processing in the pituitary, central nervous system, and peripheral tissues. Endocr Rev. 1988;9(1):159–79.CrossRefPubMed Smith AI, Funder JW. Proopiomelanocortin processing in the pituitary, central nervous system, and peripheral tissues. Endocr Rev. 1988;9(1):159–79.CrossRefPubMed
10.
go back to reference Raffin-Sanson ML, de Keyzer Y, Bertagna X. Proopiomelanocortin, a polypeptide precursor with multiple functions: from physiology to pathological conditions. Eur J Endocrinol. 2003;149(2):79–90.CrossRefPubMed Raffin-Sanson ML, de Keyzer Y, Bertagna X. Proopiomelanocortin, a polypeptide precursor with multiple functions: from physiology to pathological conditions. Eur J Endocrinol. 2003;149(2):79–90.CrossRefPubMed
11.
go back to reference Stein C, et al. Opioids from immunocytes interact with receptors on sensory nerves to inhibit nociception in inflammation. Proc Natl Acad Sci USA. 1990;87(15):5935–9.CrossRefPubMedPubMedCentral Stein C, et al. Opioids from immunocytes interact with receptors on sensory nerves to inhibit nociception in inflammation. Proc Natl Acad Sci USA. 1990;87(15):5935–9.CrossRefPubMedPubMedCentral
12.
go back to reference Challis BG, et al. Mice lacking pro-opiomelanocortin are sensitive to high-fat feeding but respond normally to the acute anorectic effects of peptide-YY(3-36). Proc Natl Acad Sci USA. 2004;101(13):4695–700.CrossRefPubMedPubMedCentral Challis BG, et al. Mice lacking pro-opiomelanocortin are sensitive to high-fat feeding but respond normally to the acute anorectic effects of peptide-YY(3-36). Proc Natl Acad Sci USA. 2004;101(13):4695–700.CrossRefPubMedPubMedCentral
13.
go back to reference Coll AP, Loraine Tung YC. Pro-opiomelanocortin (POMC)-derived peptides and the regulation of energy homeostasis. Mol Cell Endocrinol. 2009;300(1–2):147–51.CrossRefPubMed Coll AP, Loraine Tung YC. Pro-opiomelanocortin (POMC)-derived peptides and the regulation of energy homeostasis. Mol Cell Endocrinol. 2009;300(1–2):147–51.CrossRefPubMed
15.
go back to reference Lundblad JR, Roberts JL. Regulation of proopiomelanocortin gene expression in pituitary. Endocr Rev. 1988;9(1):135–58.CrossRefPubMed Lundblad JR, Roberts JL. Regulation of proopiomelanocortin gene expression in pituitary. Endocr Rev. 1988;9(1):135–58.CrossRefPubMed
16.
go back to reference Perrin MH, Vale WW. Corticotropin releasing factor receptors and their ligand family. Ann N Y Acad Sci. 1999;885:312–28.CrossRefPubMed Perrin MH, Vale WW. Corticotropin releasing factor receptors and their ligand family. Ann N Y Acad Sci. 1999;885:312–28.CrossRefPubMed
17.
go back to reference Muller MB, et al. Expression of CRHR1 and CRHR2 in mouse pituitary and adrenal gland: implications for HPA system regulation. Endocrinology. 2001;142(9):4150–3.CrossRefPubMed Muller MB, et al. Expression of CRHR1 and CRHR2 in mouse pituitary and adrenal gland: implications for HPA system regulation. Endocrinology. 2001;142(9):4150–3.CrossRefPubMed
19.
go back to reference Harden RN, et al. Validation of proposed diagnostic criteria (the “Budapest Criteria”) for Complex Regional Pain Syndrome. Pain. 2010;150(2):268–74.CrossRefPubMedPubMedCentral Harden RN, et al. Validation of proposed diagnostic criteria (the “Budapest Criteria”) for Complex Regional Pain Syndrome. Pain. 2010;150(2):268–74.CrossRefPubMedPubMedCentral
20.
go back to reference Schwartzman RJ, et al. Outpatient intravenous ketamine for the treatment of complex regional pain syndrome: a double-blind placebo controlled study. Pain. 2009;147(1–3):107–15.CrossRefPubMed Schwartzman RJ, et al. Outpatient intravenous ketamine for the treatment of complex regional pain syndrome: a double-blind placebo controlled study. Pain. 2009;147(1–3):107–15.CrossRefPubMed
21.
go back to reference Goldberg ME, et al. Pharmacodynamic profiles of ketamine (R)- and (S)- with 5-day inpatient infusion for the treatment of complex regional pain syndrome. Pain Physician. 2010;13(4):379–87.PubMedPubMedCentral Goldberg ME, et al. Pharmacodynamic profiles of ketamine (R)- and (S)- with 5-day inpatient infusion for the treatment of complex regional pain syndrome. Pain Physician. 2010;13(4):379–87.PubMedPubMedCentral
22.
go back to reference Klega A, et al. Central opioidergic neurotransmission in complex regional pain syndrome. Neurology. 2010;75(2):129–36.CrossRefPubMed Klega A, et al. Central opioidergic neurotransmission in complex regional pain syndrome. Neurology. 2010;75(2):129–36.CrossRefPubMed
23.
go back to reference Takahashi M, et al. Lower beta-endorphin content of peripheral blood mononuclear cells in patients with complex regional pain syndrome. J Back Musculoskelet Rehabil. 2000;15(1):31–6.PubMed Takahashi M, et al. Lower beta-endorphin content of peripheral blood mononuclear cells in patients with complex regional pain syndrome. J Back Musculoskelet Rehabil. 2000;15(1):31–6.PubMed
24.
go back to reference Choi SE, et al. Elevated microRNA-34a in obesity reduces NAD + levels and SIRT1 activity by directly targeting NAMPT. Aging Cell. 2013;12(6):1062–72.CrossRefPubMed Choi SE, et al. Elevated microRNA-34a in obesity reduces NAD + levels and SIRT1 activity by directly targeting NAMPT. Aging Cell. 2013;12(6):1062–72.CrossRefPubMed
25.
go back to reference Fu T, et al. Aberrantly elevated microRNA-34a in obesity attenuates hepatic responses to FGF19 by targeting a membrane coreceptor beta-Klotho. Proc Natl Acad Sci USA. 2012;109(40):16137–42.CrossRefPubMedPubMedCentral Fu T, et al. Aberrantly elevated microRNA-34a in obesity attenuates hepatic responses to FGF19 by targeting a membrane coreceptor beta-Klotho. Proc Natl Acad Sci USA. 2012;109(40):16137–42.CrossRefPubMedPubMedCentral
26.
go back to reference Azari P, et al. Efficacy and safety of ketamine in patients with complex regional pain syndrome: a systematic review. CNS Drugs. 2012;26(3):215–28.CrossRefPubMed Azari P, et al. Efficacy and safety of ketamine in patients with complex regional pain syndrome: a systematic review. CNS Drugs. 2012;26(3):215–28.CrossRefPubMed
28.
go back to reference Pritchard LE, Turnbull AV, White A. Pro-opiomelanocortin processing in the hypothalamus: impact on melanocortin signalling and obesity. J Endocrinol. 2002;172(3):411–21.CrossRefPubMed Pritchard LE, Turnbull AV, White A. Pro-opiomelanocortin processing in the hypothalamus: impact on melanocortin signalling and obesity. J Endocrinol. 2002;172(3):411–21.CrossRefPubMed
29.
go back to reference Morton GJ, et al. Central nervous system control of food intake and body weight. Nature. 2006;443(7109):289–95.CrossRefPubMed Morton GJ, et al. Central nervous system control of food intake and body weight. Nature. 2006;443(7109):289–95.CrossRefPubMed
30.
go back to reference Valassi E, Scacchi M, Cavagnini F. Neuroendocrine control of food intake. Nutr Metab Cardiovasc Dis. 2008;18(2):158–68.CrossRefPubMed Valassi E, Scacchi M, Cavagnini F. Neuroendocrine control of food intake. Nutr Metab Cardiovasc Dis. 2008;18(2):158–68.CrossRefPubMed
31.
go back to reference Blalock JE. Proopiomelanocortin and the immune-neuroendocrine connection. Ann N Y Acad Sci. 1999;885:161–72.CrossRefPubMed Blalock JE. Proopiomelanocortin and the immune-neuroendocrine connection. Ann N Y Acad Sci. 1999;885:161–72.CrossRefPubMed
32.
go back to reference Lyons PD, Blalock JE. Pro-opiomelanocortin gene expression and protein processing in rat mononuclear leukocytes. J Neuroimmunol. 1997;78(1):47–56.CrossRefPubMed Lyons PD, Blalock JE. Pro-opiomelanocortin gene expression and protein processing in rat mononuclear leukocytes. J Neuroimmunol. 1997;78(1):47–56.CrossRefPubMed
33.
go back to reference Lucas N, et al. Chronic delivery of alpha-melanocyte-stimulating hormone in rat hypothalamus using albumin-alginate microparticles: effects on food intake and body weight. Neuroscience. 2015;290:445–53.CrossRefPubMed Lucas N, et al. Chronic delivery of alpha-melanocyte-stimulating hormone in rat hypothalamus using albumin-alginate microparticles: effects on food intake and body weight. Neuroscience. 2015;290:445–53.CrossRefPubMed
34.
go back to reference Starowicz K, Przewlocka B. The role of melanocortins and their receptors in inflammatory processes, nerve regeneration and nociception. Life Sci. 2003;73(7):823–47.CrossRefPubMed Starowicz K, Przewlocka B. The role of melanocortins and their receptors in inflammatory processes, nerve regeneration and nociception. Life Sci. 2003;73(7):823–47.CrossRefPubMed
35.
go back to reference Baltazi M, et al. Plasma neuropeptide Y (NPY) and alpha-melanocyte stimulating hormone (a-MSH) levels in patients with or without hypertension and/or obesity: a pilot study. Am J Cardiovasc Dis. 2011;1(1):48–59.PubMedPubMedCentral Baltazi M, et al. Plasma neuropeptide Y (NPY) and alpha-melanocyte stimulating hormone (a-MSH) levels in patients with or without hypertension and/or obesity: a pilot study. Am J Cardiovasc Dis. 2011;1(1):48–59.PubMedPubMedCentral
36.
go back to reference Nam SY, et al. Cerebrospinal fluid and plasma concentrations of leptin, NPY, and alpha-MSH in obese women and their relationship to negative energy balance. J Clin Endocrinol Metab. 2001;86(10):4849–53.PubMed Nam SY, et al. Cerebrospinal fluid and plasma concentrations of leptin, NPY, and alpha-MSH in obese women and their relationship to negative energy balance. J Clin Endocrinol Metab. 2001;86(10):4849–53.PubMed
37.
go back to reference Hoggard N, et al. Plasma concentrations of alpha-MSH, AgRP and leptin in lean and obese men and their relationship to differing states of energy balance perturbation. Clin Endocrinol (Oxf). 2004;61(1):31–9.CrossRef Hoggard N, et al. Plasma concentrations of alpha-MSH, AgRP and leptin in lean and obese men and their relationship to differing states of energy balance perturbation. Clin Endocrinol (Oxf). 2004;61(1):31–9.CrossRef
38.
go back to reference YaDeau JT, Morelli CM, Billingsley JK. Ketamine stimulates secretion of beta-endorphin from a mouse pituitary cell line. Reg Anesth Pain Med. 2003;28(1):12–6.CrossRefPubMed YaDeau JT, Morelli CM, Billingsley JK. Ketamine stimulates secretion of beta-endorphin from a mouse pituitary cell line. Reg Anesth Pain Med. 2003;28(1):12–6.CrossRefPubMed
39.
40.
go back to reference Adams ML, et al. The role of endogenous peptides in the action of opioid analgesics. Ann Emerg Med. 1986;15(9):1030–5.CrossRefPubMed Adams ML, et al. The role of endogenous peptides in the action of opioid analgesics. Ann Emerg Med. 1986;15(9):1030–5.CrossRefPubMed
41.
go back to reference Gustin SM, et al. NMDA-receptor antagonist and morphine decrease CRPS-pain and cerebral pain representation. Pain. 2010;151(1):69–76.CrossRefPubMed Gustin SM, et al. NMDA-receptor antagonist and morphine decrease CRPS-pain and cerebral pain representation. Pain. 2010;151(1):69–76.CrossRefPubMed
42.
go back to reference Nikolarakis KE, Almeida OF, Herz A. Feedback inhibition of opioid peptide release in the hypothalamus of the rat. Neuroscience. 1987;23(1):143–8.CrossRefPubMed Nikolarakis KE, Almeida OF, Herz A. Feedback inhibition of opioid peptide release in the hypothalamus of the rat. Neuroscience. 1987;23(1):143–8.CrossRefPubMed
43.
go back to reference Garcia de Yebenes E, Pelletier G. Opioid regulation of proopiomelanocortin (POMC) gene expression in the rat brain as studied by in situ hybridization. Neuropeptides. 1993;25(2):91–4.CrossRefPubMed Garcia de Yebenes E, Pelletier G. Opioid regulation of proopiomelanocortin (POMC) gene expression in the rat brain as studied by in situ hybridization. Neuropeptides. 1993;25(2):91–4.CrossRefPubMed
44.
go back to reference Jaffe SB, Sobieszczyk S, Wardlaw SL. Effect of opioid antagonism on beta-endorphin processing and proopiomelanocortin-peptide release in the hypothalamus. Brain Res. 1994;648(1):24–31.CrossRefPubMed Jaffe SB, Sobieszczyk S, Wardlaw SL. Effect of opioid antagonism on beta-endorphin processing and proopiomelanocortin-peptide release in the hypothalamus. Brain Res. 1994;648(1):24–31.CrossRefPubMed
45.
go back to reference Wardlaw SL, Kim J, Sobieszczyk S. Effect of morphine on proopiomelanocortin gene expression and peptide levels in the hypothalamus. Brain Res Mol Brain Res. 1996;41(1–2):140–7.CrossRefPubMed Wardlaw SL, Kim J, Sobieszczyk S. Effect of morphine on proopiomelanocortin gene expression and peptide levels in the hypothalamus. Brain Res Mol Brain Res. 1996;41(1–2):140–7.CrossRefPubMed
46.
go back to reference Affolter HU, Reisine T. Corticotropin releasing factor increases proopiomelanocortin messenger RNA in mouse anterior pituitary tumor cells. J Biol Chem. 1985;260(29):15477–81.PubMed Affolter HU, Reisine T. Corticotropin releasing factor increases proopiomelanocortin messenger RNA in mouse anterior pituitary tumor cells. J Biol Chem. 1985;260(29):15477–81.PubMed
47.
go back to reference Agelaki S, et al. Corticotropin-releasing hormone augments proinflammatory cytokine production from macrophages in vitro and in lipopolysaccharide-induced endotoxin shock in mice. Infect Immun. 2002;70(11):6068–74.CrossRefPubMedPubMedCentral Agelaki S, et al. Corticotropin-releasing hormone augments proinflammatory cytokine production from macrophages in vitro and in lipopolysaccharide-induced endotoxin shock in mice. Infect Immun. 2002;70(11):6068–74.CrossRefPubMedPubMedCentral
48.
go back to reference Webster EL, et al. Corticotropin releasing hormone (CRH) antagonist attenuates adjuvant induced arthritis: role of CRH in peripheral inflammation. J Rheumatol. 2002;29(6):1252–61.PubMed Webster EL, et al. Corticotropin releasing hormone (CRH) antagonist attenuates adjuvant induced arthritis: role of CRH in peripheral inflammation. J Rheumatol. 2002;29(6):1252–61.PubMed
49.
go back to reference De Guire V, et al. Circulating miRNAs as sensitive and specific biomarkers for the diagnosis and monitoring of human diseases: promises and challenges. Clin Biochem. 2013;46(10–11):846–60.CrossRefPubMed De Guire V, et al. Circulating miRNAs as sensitive and specific biomarkers for the diagnosis and monitoring of human diseases: promises and challenges. Clin Biochem. 2013;46(10–11):846–60.CrossRefPubMed
Metadata
Title
Hsa-miR-34a mediated repression of corticotrophin releasing hormone receptor 1 regulates pro-opiomelanocortin expression in patients with complex regional pain syndrome
Authors
Botros B. Shenoda
Guillermo M. Alexander
Seena K. Ajit
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2016
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-016-0820-1

Other articles of this Issue 1/2016

Journal of Translational Medicine 1/2016 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine