51
|
Kumar S, Ruchi R, James SR, Chidiac EJ. Gene therapy for chronic neuropathic pain: how does it work and where do we stand today? PAIN MEDICINE 2011; 12:808-22. [PMID: 21564510 DOI: 10.1111/j.1526-4637.2011.01120.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Chronic neuropathic pain has been an enigma to physicians and researchers for decades. A better understanding of its pathophysiology has given us more insight into its various mechanisms and possible treatment options. We now have an understanding of the role of various ionic channels, biologically active molecules involved in pain, and also the intricate pain pathways where possible interventions might lead to substantial pain relief. The recent research on laboratory animals using virus-based vectors for gene transfer at targeted sites is very promising and may lead to additional human clinical trials. However, one needs to be aware that this "novel" approach is still in its infancy and that many of its details need to be further elucidated. The purpose of this article is to thoroughly review the current available literature and analyze the deficiencies in our current knowledge. DESIGN Literature review. METHODS After an extensive online literature search, a total of 133 articles were selected to synthesize a comprehensive review about chronic neuropathic pain and gene therapy in order to understand the concepts and mechanisms. RESULTS Most of the studies have shown benefits of gene therapy in animal models, and recently, phase 1 human trials using herpes simplex virus vector have started for intractable cancer pain. CONCLUSION Although animal data have shown safety and efficacy, and initial human trials have been promising, additional studies in humans are required to more completely understand the actual benefits and risks of using gene therapy for the treatment of chronic neuropathic pain.
Collapse
Affiliation(s)
- Sanjeev Kumar
- Department of Anesthesiology, Wayne State University/Detroit Medical Center, Harper University Hospital, MI 48201, USA
| | | | | | | |
Collapse
|
52
|
Handy CR, Krudy C, Boulis N. Gene therapy: a potential approach for cancer pain. PAIN RESEARCH AND TREATMENT 2011; 2011:987597. [PMID: 22110939 PMCID: PMC3196247 DOI: 10.1155/2011/987597] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 12/14/2010] [Accepted: 01/21/2011] [Indexed: 12/21/2022]
Abstract
Chronic pain is experienced by as many as 90% of cancer patients at some point during the disease. This pain can be directly cancer related or arise from a sensory neuropathy related to chemotherapy. Major pharmacological agents used to treat cancer pain often lack anatomical specificity and can have off-target effects that create new sources of suffering. These concerns establish a need for improved cancer pain management. Gene therapy is emerging as an exciting prospect. This paper discusses the potential for viral vector-based treatment of cancer pain. It describes studies involving vector delivery of transgenes to laboratory pain models to modulate the nociceptive cascade. It also discusses clinical investigations aimed at regulating pain in cancer patients. Considering the prevalence of pain among cancer patients and the growing potential of gene therapy, these studies could set the stage for a new class of medicines that selectively disrupt nociceptive signaling with limited off-target effects.
Collapse
Affiliation(s)
- Chalonda R. Handy
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Rm 6339, Atlanta, GA 30322, USA
| | - Christina Krudy
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Rm 6339, Atlanta, GA 30322, USA
| | - Nicholas Boulis
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Rm 6339, Atlanta, GA 30322, USA
| |
Collapse
|
53
|
Sun J, Liu S, Mata M, Fink DJ, Hao S. Transgene-mediated expression of tumor necrosis factor soluble receptor attenuates morphine tolerance in rats. Gene Ther 2011; 19:101-8. [PMID: 21614028 PMCID: PMC3175012 DOI: 10.1038/gt.2011.76] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Opiate/narcotic analgesics are the most effective treatments for chronic severe pain, but their clinical utility is often hampered by the development of analgesic tolerance. Recent evidence suggests chronic morphine may activate glial cells to release proinflammatory cytokines. In this study, we used herpes simplex virus (HSV) vectors-based gene transfer to dorsal root ganglion to produce a local release of p55 TNF soluble receptor in the spinal cord in rats with morphine tolerance. Subcutaneous inoculation of HSV vectors expressing p55 TNF soluble receptor into the plantar surface of the hindpaws, enhanced the antinociceptive effect of acute morphine in rats. Subcutaneous inoculation of those vectors into hindpaws also delayed the development of chronic morphine tolerance in rats. TNF soluble receptor expressed by HSV vector reduced gene transcription of mRNA of spinal TNFα and IL-1β induced by repeated morphine. Furthermore, we found that TNF soluble receptor mediated by HSV, reversed the upregulation of TNFα, IL-1β and phosphorylation of p38 mitogen-activated protein kinase (MAPK) induced by repeated morphine. These results support the concept that proinflammatory cytokines may play an important role in the pathogenesis induced by morphine. This study provides a novel approach to treating morphine tolerance.
Collapse
Affiliation(s)
- J Sun
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | |
Collapse
|
54
|
Zheng W, Ouyang H, Zheng X, Liu S, Mata M, Fink DJ, Hao S. Glial TNFα in the spinal cord regulates neuropathic pain induced by HIV gp120 application in rats. Mol Pain 2011; 7:40. [PMID: 21599974 PMCID: PMC3121595 DOI: 10.1186/1744-8069-7-40] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 05/20/2011] [Indexed: 01/21/2023] Open
Abstract
Background HIV-associated sensory neuropathy (HIV-SN) is one of the most common forms of peripheral neuropathy, affecting about 30% of people with acquired immune deficiency syndrome (AIDS). The symptoms of HIV-SN are dominated by neuropathic pain. Glia activation in the spinal cord has become an attractive target for attenuating chronic pain. This study will investigate the role of spinal TNFα released from glia in HIV-related neuropathic pain. Results Peripheral gp120 application into the rat sciatic nerve induced mechanical allodynia for more than 7 weeks, and upregulated the expression of spinal TNFα in the mRNA and the protein levels at 2 weeks after gp120 application. Spinal TNFα was colocalized with GFAP (a marker of astrocytes) and Iba1 (a marker of microglia) in immunostaining, suggesting that glia produce TNFα in the spinal cord in this model. Peripheral gp120 application also increased TNFα in the L4/5 DRG. Furthermore, intrathecal administration of TNFα siRNA or soluble TNF receptor reduced gp120 application-induced mechanical allodynia. Conclusions Our results indicate that TNFα in the spinal cord and the DRG are involved in neuropathic pain, following the peripheral HIV gp120 application, and that blockade of the glial product TNFα reverses neuropathic pain induced by HIV gp120 application.
Collapse
Affiliation(s)
- Wenwen Zheng
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | |
Collapse
|
55
|
Hao S, Liu S, Zheng X, Zheng W, Ouyang H, Mata M, Fink DJ. The role of TNFα in the periaqueductal gray during naloxone-precipitated morphine withdrawal in rats. Neuropsychopharmacology 2011; 36:664-76. [PMID: 21068718 PMCID: PMC3055683 DOI: 10.1038/npp.2010.197] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tolerance and dependence are common complications of long-term treatment of pain with opioids, which substantially limit the long-term use of these drugs. The mechanisms underlying these phenomena are poorly understood. Studies have implicated the midbrain periaqueductal gray (PAG) in the pathogenesis of morphine withdrawal, and recent evidence suggests that proinflammatory cytokines in the PAG may play an important role in morphine withdrawal. Here we report that chronic morphine withdrawal-induced upregulation of glial fibrillary acidic protein (GFAP), tumor necrosis factor alpha (TNFα) and phosphorylation of ERK1/2 (pERK1/2) in the caudal ventrolateral PAG (vlPAG). Microinjection of recombinant TNFα into the vlPAG followed by intraperitoneal naloxone resulted in morphine withdrawal-like behavioral signs, and upregulation of pERK1/2, expression of Fos, and phosphorylation of cAMP response element binding (pCREB) protein. We used a herpes simplex virus (HSV)-based vector expressing p55 soluble TNF receptor (sTNFR) microinjected into the PAG to examine the role of the proinflammatory cytokine TNFα in the PAG in the naloxone-precipitated withdrawal response. Microinjection of HSV vector expressing sTNFR into the PAG before the start of morphine treatment significantly reduced the naloxone-precipitated withdrawal behavioral response and downregulated the expression of GFAP and TNFα in astrocytes of the PAG. TNFR type I colocalized with neuronal pERK1/2. Microinjection of HSV vector expressing sTNFR into the PAG also significantly reduced the phosphorylation of both ERK1/2 and CREB, and reduced Fos immunoreactivity in neurons of the PAG following naloxone-precipitated withdrawal. These results support the concept that proinflammatory cytokines expressed in astrocytes in the PAG may play an important role in the pathogenesis of morphine withdrawal response.
Collapse
Affiliation(s)
- Shuanglin Hao
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| | - Shue Liu
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA,Ann Arbor VA Healthcare System, Ann Arbor, MI, USA
| | - Xuexing Zheng
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA,Ann Arbor VA Healthcare System, Ann Arbor, MI, USA
| | - Wenwen Zheng
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA,Ann Arbor VA Healthcare System, Ann Arbor, MI, USA
| | - Handong Ouyang
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA,Ann Arbor VA Healthcare System, Ann Arbor, MI, USA
| | - Marina Mata
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA,Ann Arbor VA Healthcare System, Ann Arbor, MI, USA
| | - David J Fink
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA,Ann Arbor VA Healthcare System, Ann Arbor, MI, USA
| |
Collapse
|
56
|
TNF-α contributes to spinal cord synaptic plasticity and inflammatory pain: distinct role of TNF receptor subtypes 1 and 2. Pain 2010; 152:419-427. [PMID: 21159431 DOI: 10.1016/j.pain.2010.11.014] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 10/19/2010] [Accepted: 11/16/2010] [Indexed: 12/30/2022]
Abstract
Tumor necrosis factor-alpha (TNF-α) is a key proinflammatory cytokine. It is generally believed that TNF-α exerts its effects primarily via TNF receptor subtype-1 (TNFR1). We investigated the distinct roles of TNFR1 and TNFR2 in spinal cord synaptic transmission and inflammatory pain. Compared to wild-type (WT) mice, TNFR1- and TNFR2-knockout (KO) mice exhibited normal heat sensitivity and unaltered excitatory synaptic transmission in the spinal cord, as revealed by spontaneous excitatory postsynaptic currents in lamina II neurons of spinal cord slices. However, heat hyperalgesia after intrathecal TNF-α and the second-phase spontaneous pain in the formalin test were reduced in both TNFR1- and TNFR2-KO mice. In particular, heat hyperalgesia after intraplantar injection of complete Freund's adjuvant (CFA) was decreased in the early phase in TNFR2-KO mice but reduced in both the early and later phase in TNFR1-KO mice. Consistently, CFA elicited a transient increase of TNFR2 mRNA levels in the spinal cord on day 1. Notably, TNF-α evoked a drastic increase in spontaneous excitatory postsynaptic current frequency in lamina II neurons, which was abolished in TNFR1-KO mice and reduced in TNFR2-KO mice. TNF-α also increased N-methyl-d-aspartate (NMDA) currents in lamina II neurons, and this increase was abolished in TNFR1-KO mice but retained in TNFR2-KO mice. Finally, intrathecal injection of the NMDA receptor antagonist MK-801 prevented heat hyperalgesia elicited by intrathecal TNF-α. Our findings support a central role of TNF-α in regulating synaptic plasticity (central sensitization) and inflammatory pain via both TNFR1 and TNFR2. Our data also uncover a unique role of TNFR2 in mediating early-phase inflammatory pain. TNF-α is shown to play a critical role in regulating spinal cord synaptic plasticity and central sensitization, and TNFR1 and TNFR2 play a distinct role in regulating different phases of inflammatory pain.
Collapse
|
57
|
HSV delivery of a ligand-regulated endogenous ion channel gene to sensory neurons results in pain control following channel activation. Mol Ther 2010; 19:500-6. [PMID: 21081904 DOI: 10.1038/mt.2010.246] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Persistent pain remains a tremendous health problem due to both its prevalence and dearth of effective therapeutic interventions. To maximize pain relief while minimizing side effects, current gene therapy-based approaches have mostly exploited the expression of pain inhibitory products or interfered with pronociceptive ion channels. These methods do not enable control over the timing or duration of analgesia, nor titration to analgesic efficacy. Here, we describe a gene therapy strategy that potentially overcomes these limitations by providing exquisite control over therapy with efficacy in clinically relevant models of inflammatory pain. We utilize a herpes simplex viral (HSV) vector (vHGlyRα1) to express a ligand-regulated chloride ion channel, the glycine receptor (GlyR) in targeted sensory afferents; the subsequent exogenous addition of glycine provides the means for temporal and spatial control of afferent activity, and therefore pain. Use of an endogenous inhibitory receptor not normally present on sensory neurons both minimizes immunogenicity and maximizes therapeutic selectivity.
Collapse
|
58
|
Zhou Z, Peng X, Hagshenas J, Insolera R, Fink DJ, Mata M. A novel cell-cell signaling by microglial transmembrane TNFα with implications for neuropathic pain. Pain 2010; 151:296-306. [PMID: 20609516 DOI: 10.1016/j.pain.2010.06.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 06/03/2010] [Accepted: 06/15/2010] [Indexed: 02/01/2023]
Abstract
Neuropathic pain is accompanied by neuroimmune activation in dorsal horn of spinal cord. We have observed that in animal models this activation is characterized by an increased expression of transmembrane tumor necrosis factor α (mTNFα) without the release of soluble tumor necrosis factor α (sTNFα). Herein we report that the pain-related neurotransmitter peptide substance P (SP) increases the expression of mTNFα without the release of sTNFα from primary microglial cells. We modeled this interaction using an immortalized microglial cell line; exposure of these cells to SP also resulted in the increased expression of mTNFα but without any increase in the expression of the TNF-cleaving enzyme (TACE) and no release of sTNFα. In order to evaluate the biological function of uncleaved mTNFα, we transfected COS-7 cells with a mutant full-length TNFα construct resistant to cleavage by TACE. Coculture of COS-7 cells expressing the mutant TNFα with microglial cells led to microglial cell activation indicated by increased OX42 immunoreactivity and release of macrophage chemoattractant peptide 1 (CCL2) by direct cell-cell contact. These results suggest a novel pathway through which the release of SP by primary afferents activates microglial expression of mTNFα, establishing a feed-forward loop that may contribute to the establishment of chronic pain.
Collapse
Affiliation(s)
- Zhigang Zhou
- Department of Neurology, University of Michigan, USA VA Ann Arbor Healthcare System, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
59
|
Abstract
The very deep knowledge acquired on the genetics and molecular biology of herpes simplex virus (HSV), has allowed the development of potential replication-competent and replication-defective vectors for several applications in human healthcare. These include delivery and expression of human genes to cells of the nervous systems, selective destruction of cancer cells, prophylaxis against infection with HSV or other infectious diseases, and targeted infection to specific tissues or organs. Replication-defective recombinant vectors are non-toxic gene transfer tools that preserve most of the neurotropic features of wild type HSV-1, particularly the ability to express genes after having established latent infections, and are thus proficient candidates for therapeutic gene transfer settings in neurons. A replication-defective HSV vector for the treatment of pain has recently entered in phase 1 clinical trial. Replication-competent (oncolytic) vectors are becoming a suitable and powerful tool to eradicate brain tumours due to their ability to replicate and spread only within the tumour mass, and have reached phase II/III clinical trials in some cases. The progress in understanding the host immune response induced by the vector is also improving the use of HSV as a vaccine vector against both HSV infection and other pathogens. This review briefly summarizes the obstacle encountered in the delivery of HSV vectors and examines the various strategies developed or proposed to overcome such challenges.
Collapse
Affiliation(s)
- Roberto Manservigi
- Department of Experimental and Diagnostic Medicine - Section of Microbiology, University of Ferrara, Via Luigi Borsari 46, 44100 Ferrara, Italy
| | | | | |
Collapse
|
60
|
Manservigi R, Argnani R, Marconi P. HSV Recombinant Vectors for Gene Therapy. Open Virol J 2010; 4:123-56. [PMID: 20835362 DOI: 10.2174/1874357901004030123] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 03/13/2010] [Accepted: 03/31/2010] [Indexed: 12/16/2022] Open
Abstract
The very deep knowledge acquired on the genetics and molecular biology of herpes simplex virus (HSV), has allowed the development of potential replication-competent and replication-defective vectors for several applications in human healthcare. These include delivery and expression of human genes to cells of the nervous systems, selective destruction of cancer cells, prophylaxis against infection with HSV or other infectious diseases, and targeted infection to specific tissues or organs. Replication-defective recombinant vectors are non-toxic gene transfer tools that preserve most of the neurotropic features of wild type HSV-1, particularly the ability to express genes after having established latent infections, and are thus proficient candidates for therapeutic gene transfer settings in neurons. A replication-defective HSV vector for the treatment of pain has recently entered in phase 1 clinical trial. Replication-competent (oncolytic) vectors are becoming a suitable and powerful tool to eradicate brain tumours due to their ability to replicate and spread only within the tumour mass, and have reached phase II/III clinical trials in some cases. The progress in understanding the host immune response induced by the vector is also improving the use of HSV as a vaccine vector against both HSV infection and other pathogens. This review briefly summarizes the obstacle encountered in the delivery of HSV vectors and examines the various strategies developed or proposed to overcome such challenges.
Collapse
Affiliation(s)
- Roberto Manservigi
- Department of Experimental and Diagnostic Medicine - Section of Microbiology, University of Ferrara, Via Luigi Borsari 46, 44100 Ferrara, Italy
| | | | | |
Collapse
|
61
|
Leung L, Cahill CM. TNF-alpha and neuropathic pain--a review. J Neuroinflammation 2010; 7:27. [PMID: 20398373 PMCID: PMC2861665 DOI: 10.1186/1742-2094-7-27] [Citation(s) in RCA: 436] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Accepted: 04/16/2010] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor alpha (TNF-α) was discovered more than a century ago, and its known roles have extended from within the immune system to include a neuro-inflammatory domain in the nervous system. Neuropathic pain is a recognized type of pathological pain where nociceptive responses persist beyond the resolution of damage to the nerve or its surrounding tissue. Very often, neuropathic pain is disproportionately enhanced in intensity (hyperalgesia) or altered in modality (hyperpathia or allodynia) in relation to the stimuli. At time of this writing, there is as yet no common consensus about the etiology of neuropathic pain - possible mechanisms can be categorized into peripheral sensitization and central sensitization of the nervous system in response to the nociceptive stimuli. Animal models of neuropathic pain based on various types of nerve injuries (peripheral versus spinal nerve, ligation versus chronic constrictive injury) have persistently implicated a pivotal role for TNF-α at both peripheral and central levels of sensitization. Despite a lack of success in clinical trials of anti-TNF-α therapy in alleviating the sciatic type of neuropathic pain, the intricate link of TNF-α with other neuro-inflammatory signaling systems (e.g., chemokines and p38 MAPK) has indeed inspired a systems approach perspective for future drug development in treating neuropathic pain.
Collapse
Affiliation(s)
- Lawrence Leung
- Centre for Neurosciences Studies, 18, Stuart Street, Queen's University, Kingston, Ontario K7L 3N6, Canada.
| | | |
Collapse
|
62
|
HSV vector-mediated modification of primary nociceptor afferents: an approach to inhibit chronic pain. Gene Ther 2010; 16:493-501. [PMID: 19357694 DOI: 10.1038/gt.2009.24] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic pain is a serious medical condition with millions of sufferers for whom long-term therapies are either lacking or inadequate. Here we review the use of herpes simplex virus vectors as therapeutic tools to treat chronic pain by gene therapy. We describe an approach to inhibit chronic pain signaling whereby vector-mediated genes transferred to sensory nerves will modify the primary afferent nociceptor to prevent pain signaling to second-order nerves in the spinal cord. This approach may be used to reverse the chronic pain state of the nociceptor and could affect downstream pain-related changes in the central nervous system.
Collapse
|
63
|
Wolfe D, Wechuck J, Krisky D, Mata M, Fink DJ. A clinical trial of gene therapy for chronic pain. PAIN MEDICINE 2010; 10:1325-30. [PMID: 19818042 DOI: 10.1111/j.1526-4637.2009.00720.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The first human trial of gene therapy for chronic pain, a phase 1 study of a nonreplicating herpes simplex virus (HSV)-based vector engineered to express preproenkephalin in patients with intractable pain from cancer, began enrolling subjects in December 2008. In this article, we describe the rationale underlying this potential approach to treatment of pain, the preclinical animal data in support of this approach, the design of the study, and studies with additional HSV-based vectors that may be used to develop treatment for other types of pain.
Collapse
|
64
|
Kim SJ, Lee WI, Lee YS, Kim DH, Chang JW, Kim SW, Lee H. Effective relief of neuropathic pain by adeno-associated virus-mediated expression of a small hairpin RNA against GTP cyclohydrolase 1. Mol Pain 2009; 5:67. [PMID: 19922668 PMCID: PMC2785765 DOI: 10.1186/1744-8069-5-67] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 11/18/2009] [Indexed: 12/30/2022] Open
Abstract
Background Recent studies show that transcriptional activation of GTP cyclohydrolase I (GCH1) in dorsal root ganglia (DRG) is significantly involved in the development and persistency of pain symptoms. We thus hypothesize that neuropathic pain may be attenuated by down-regulation of GCH1 expression, and propose a gene silencing system for this purpose. Results To interrupt GCH1 synthesis, we designed a bidirectional recombinant adeno-associated virus encoding both a small hairpin RNA against GCH1 and a GFP reporter gene (rAAV-shGCH1). After rAAV-shGCH1 was introduced into the sciatic nerve prior to or following pain-inducing surgery, therapeutic efficacy and the underlying mechanisms were subsequently validated in animal models. The GFP expression data indicates that rAAV effectively delivered transgenes to DRG. Subsequently reduced GCH1 expression was evident from immunohistochemistry and western-blotting analysis. Along with the down-regulation of GCH1, the von Frey test correspondingly indicated a sharp decline in pain symptoms upon both pre- and post-treatment with rAAV-shGCH1. Interestingly, GCH1 down-regulation additionally led to decreased microglial activation in the dorsal horn, implying an association between pain attenuation and reduced inflammation. Conclusion Therefore, the data suggests that GCH1 levels can be reduced by introducing rAAV-shGCH1, leading to pain relief. Based on the results, we propose that GCH1 modulation may be developed as a clinically applicable gene therapy strategy to treat neuropathic pain.
Collapse
Affiliation(s)
- Sung Jin Kim
- Departments of Microbiology, University of Ulsan College of Medicine, Seoul, Korea.
| | | | | | | | | | | | | |
Collapse
|
65
|
Towne C, Pertin M, Beggah AT, Aebischer P, Decosterd I. Recombinant adeno-associated virus serotype 6 (rAAV2/6)-mediated gene transfer to nociceptive neurons through different routes of delivery. Mol Pain 2009; 5:52. [PMID: 19737386 PMCID: PMC2747840 DOI: 10.1186/1744-8069-5-52] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Accepted: 09/08/2009] [Indexed: 11/27/2022] Open
Abstract
Background Gene transfer to nociceptive neurons of the dorsal root ganglia (DRG) is a promising approach to dissect mechanisms of pain in rodents and is a potential therapeutic strategy for the treatment of persistent pain disorders such as neuropathic pain. A number of studies have demonstrated transduction of DRG neurons using herpes simplex virus, adenovirus and more recently, adeno-associated virus (AAV). Recombinant AAV are currently the gene transfer vehicles of choice for the nervous system and have several advantages over other vectors, including stable and safe gene expression. We have explored the capacity of recombinant AAV serotype 6 (rAAV2/6) to deliver genes to DRG neurons and characterized the transduction of nociceptors through five different routes of administration in mice. Results Direct injection of rAAV2/6 expressing green fluorescent protein (eGFP) into the sciatic nerve resulted in transduction of up to 30% eGFP-positive cells of L4 DRG neurons in a dose dependant manner. More than 90% of transduced cells were small and medium sized neurons (< 700 μm2), predominantly colocalized with markers of nociceptive neurons, and had eGFP-positive central terminal fibers in the superficial lamina of the spinal cord dorsal horn. The efficiency and profile of transduction was independent of mouse genetic background. Intrathecal administration of rAAV2/6 gave the highest level of transduction (≈ 60%) and had a similar size profile and colocalization with nociceptive neurons. Intrathecal administration also transduced DRG neurons at cervical and thoracic levels and resulted in comparable levels of transduction in a mouse model for neuropathic pain. Subcutaneous and intramuscular delivery resulted in low levels of transduction in the L4 DRG. Likewise, delivery via tail vein injection resulted in relatively few eGFP-positive cells within the DRG, however, this transduction was observed at all vertebral levels and corresponded to large non-nociceptive cell types. Conclusion We have found that rAAV2/6 is an efficient vector to deliver transgenes to nociceptive neurons in mice. Furthermore, the characterization of the transduction profile may facilitate gene transfer studies to dissect mechanisms behind neuropathic pain.
Collapse
Affiliation(s)
- Chris Towne
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, EPFL, Lausanne, Switzerland.
| | | | | | | | | |
Collapse
|
66
|
Alexander JK, DeVries AC, Kigerl KA, Dahlman JM, Popovich PG. Stress exacerbates neuropathic pain via glucocorticoid and NMDA receptor activation. Brain Behav Immun 2009; 23:851-60. [PMID: 19361551 PMCID: PMC2735409 DOI: 10.1016/j.bbi.2009.04.001] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2009] [Revised: 03/31/2009] [Accepted: 04/01/2009] [Indexed: 12/22/2022] Open
Abstract
There is growing recognition that psychological stress influences pain. Hormones that comprise the physiological response to stress (e.g., corticosterone; CORT) may interact with effectors of neuropathic pain. To test this hypothesis, mice received a spared nerve injury (SNI) after exposure to 60 min restraint stress. In stressed mice, allodynia was consistently increased. The mechanism(s) underlying the exacerbated pain response involves CORT acting via glucocorticoid receptors (GRs); RU486, a GR antagonist, prevented the stress-induced increase in allodynia whereas exogenous administration of CORT to non-stressed mice reproduced the allodynic response caused by stress. Since nerve injury-induced microglial activation has been implicated in the onset and propagation of neuropathic pain, we evaluated cellular and molecular indices of microglial activation in the context of stress. Activation of dorsal horn microglia was accelerated by stress; however, this effect was transient and was not associated with the onset or maintenance of a pro-inflammatory phenotype. Stress-enhanced allodynia was associated with increased dorsal horn extracellular signal-regulated kinase phosphorylation (pERK). ERK activation could indicate a stress-mediated increase in glutamatergic signaling, therefore mice were treated prior to SNI and stress with memantine, an N-methyl-D-aspartate receptor (NMDAR) antagonist. Memantine prevented stress-induced enhancement of allodynia after SNI. These data suggest that the hormonal responses elicited by stress exacerbate neuropathic pain through enhanced central sensitization. Moreover, drugs that inhibit glucocorticoids (GCs) and/or NMDAR signaling could ameliorate pain syndromes caused by stress.
Collapse
Affiliation(s)
- Jessica K Alexander
- Neuroscience Graduate Studies Program, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
67
|
Abstract
The numb chin syndrome (NCS) is characterized by facial numbness along the distribution of the mental branch of the trigeminal nerve. Most cases of this syndrome that are not dental in origin have been associated with malignant tumors or diffuse metastatic disease, particularly with underlying lymphoproliferative diseases and breast cancer. NCS can appear together with other signs of neoplastic dissemination or constitute the presenting symptom of the disease. The appearance of this mental nerve neuropathy should be considered as a significant symptom for clinicians, and investigations to detect a possible cancer should be mandatory. We report 12 patients with the NCS as the presenting and isolated symptom of a generalized malignancy.
Collapse
|
68
|
Maximum-entropy network analysis reveals a role for tumor necrosis factor in peripheral nerve development and function. Proc Natl Acad Sci U S A 2009; 106:12494-9. [PMID: 19597151 DOI: 10.1073/pnas.0902237106] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Gene regulatory interactions that shape developmental processes can often can be inferred from microarray analysis of gene expression, but most computational methods used require extensive datasets that can be difficult to generate. Here, we show that maximum-entropy network analysis allows extraction of genetic interactions from limited microarray datasets. Maximum-entropy networks indicated that the inflammatory cytokine TNF-alpha plays a pivotal role in Schwann cell-axon interactions, and these data suggested that TNF mediates its effects by orchestrating cytoplasmic movement and axon guidance. In vivo and in vitro experiments confirmed these predictions, showing that Schwann cells in TNF(-/-) peripheral sensory bundles fail to envelop axons efficiently, and that recombinant TNF can partially correct these defects. These data demonstrate the power of maximum-entropy network-based methods for analysis of microarray data, and they indicate that TNF-alpha plays a direct role in Schwann cell-axon communication.
Collapse
|
69
|
Niu YL, Guo Z, Zhou RH. Up-regulation of TNF-alpha in neurons of dorsal root ganglia and spinal cord during coronary artery occlusion in rats. Cytokine 2009; 47:23-9. [PMID: 19398208 DOI: 10.1016/j.cyto.2009.04.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 03/22/2009] [Accepted: 04/01/2009] [Indexed: 11/16/2022]
Abstract
Knowledge about the physiologic and pathophysiologic roles of tumor necrosis factor-alpha (TNF-alpha) in acute myocardial ischemia/infarction is still very limited. Evidence implies that TNF-alpha is involved in neural activity including nociception in peripheral and central nervous system. Current study was designed to examine the association of change in TNF-alpha and its mRNA in upper thoracic dorsal root ganglia and spinal cord (T1-T5) during acute myocardial ischemia/infarction induced by coronary artery occlusion (CAO) in rats. The experiment was performed using immunohistochemistry, enzyme immunoassay, in situ hybridization and real time reverse transcription-polymerase chain reaction techniques. At 0.5h, 1h, 3h and 6h of acute myocardial ischemia/infarction, TNF-alpha was mainly up-regulated in a sub-population of small and medium neurons and satellite cells in the dorsal root ganglia (DRG) and spinal neurons, mainly in laminae I, II and V, VI of the spinal dorsal horn of upper thoracic segments. The up-regulation of TNF-alpha mRNA was observed at 30min of CAO, which was statistically significant, compared with the control and the sham surgery groups (P<0.01). The TNF-alpha mRNA was located in the satellite cells and afferent neurons of the DRG and spinal neurons, located mainly in laminae II-VI. The findings indicate an association of up-regulation of TNF-alpha in DRG and spinal cord with acute myocardial ischemia/infarction, suggesting that TNF-alpha may be associated with the nociception initiated by acute myocardial ischemia/infarction, while the pathophysiological role needs to be studied.
Collapse
Affiliation(s)
- Yan-Lan Niu
- Department of Anesthesiology, Shanxi Medical University and Second Hospital of Shanxi Medical University, 56 Xinjian Nan Road, Taiyuan 030001, Shanxi, PR China
| | | | | |
Collapse
|
70
|
JNK-induced MCP-1 production in spinal cord astrocytes contributes to central sensitization and neuropathic pain. J Neurosci 2009; 29:4096-108. [PMID: 19339605 DOI: 10.1523/jneurosci.3623-08.2009] [Citation(s) in RCA: 455] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Our previous study showed that activation of c-jun-N-terminal kinase (JNK) in spinal astrocytes plays an important role in neuropathic pain sensitization. We further investigated how JNK regulates neuropathic pain. In cultured astrocytes, tumor necrosis factor alpha (TNF-alpha) transiently activated JNK via TNF receptor-1. Cytokine array indicated that the chemokine CCL2/MCP-1 (monocyte chemoattractant protein-1) was strongly induced by the TNF-alpha/JNK pathway. MCP-1 upregulation by TNF-alpha was dose dependently inhibited by the JNK inhibitors SP600125 (anthra[1,9-cd]pyrazol-6(2H)-one) and D-JNKI-1. Spinal injection of TNF-alpha produced JNK-dependent pain hypersensitivity and MCP-1 upregulation in the spinal cord. Furthermore, spinal nerve ligation (SNL) induced persistent neuropathic pain and MCP-1 upregulation in the spinal cord, and both were suppressed by D-JNKI-1. Remarkably, MCP-1 was primarily induced in spinal cord astrocytes after SNL. Spinal administration of MCP-1 neutralizing antibody attenuated neuropathic pain. Conversely, spinal application of MCP-1 induced heat hyperalgesia and phosphorylation of extracellular signal-regulated kinase in superficial spinal cord dorsal horn neurons, indicative of central sensitization (hyperactivity of dorsal horn neurons). Patch-clamp recordings in lamina II neurons of isolated spinal cord slices showed that MCP-1 not only enhanced spontaneous EPSCs but also potentiated NMDA- and AMPA-induced currents. Finally, the MCP-1 receptor CCR2 was expressed in neurons and some non-neuronal cells in the spinal cord. Together, we have revealed a previously unknown mechanism of MCP-1 induction and action. MCP-1 induction in astrocytes after JNK activation contributes to central sensitization and neuropathic pain facilitation by enhancing excitatory synaptic transmission. Inhibition of the JNK/MCP-1 pathway may provide a new therapy for neuropathic pain management.
Collapse
|
71
|
Grant KG, Krisky DM, Ataai MM, Glorioso JC. Engineering cell lines for production of replication defective HSV-1 gene therapy vectors. Biotechnol Bioeng 2009; 102:1087-97. [DOI: 10.1002/bit.22123] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
72
|
Abstract
Gene transfer to the dorsal root ganglion using replication defective herpes simplex virus (HSV)-based vectors reduces pain-related behaviors in rodent models having inflammatory pain, neuropathic pain and pain caused by cancer in bone. HSV vectors engineered to produce inhibitory neurotransmitters, including the delta opioid agonist peptide enkephalin, the mu opioid agonist peptide endomorphin-2 and glutamic acid decarboxylase (GAD), to effect the release of gamma amino butyric acid (GABA) act to inhibit nociceptive neurotransmission at the first synapse between primary nociceptive and second-order neuron in the dorsal horn of the spinal cord. HSV vectors engineered to release anti-inflammatory peptides, including interleukin (IL)-4, IL-10 and the p55 soluble tumor necrosis factor alpha (TNFalpha) receptor reduce neuroimmune activation in the spinal dorsal horn. The path leading from preclinical animal studies to the ongoing phase 1 human trial of the enkephalin-producing vector in patients with pain from cancer, and plans for an efficacy trial with an opioid-producing vector in inflammatory pain and an efficacy trial with a GAD-producing vector in diabetic neuropathic pain are outlined.
Collapse
|
73
|
Abstract
BACKGROUND Management of chronic pain remains a challenge in spite of the numerous drugs either approved or still in development. Apart from inadequacy of relief, there are concerns about adverse effects and addiction in the case of drugs such as opioids. Gene therapy is being investigated for improving management of pain. OBJECTIVE To addresses the rationale of gene therapy for treatment of pain and its advantages over drugs. The prospects of translation of these techniques from experimental animals to clinical use are discussed. METHODS The review is based on the available literature and is confined to experimental work, as there are no approved therapies in this category. RESULTS/CONCLUSION A number of promising gene therapies as well as antisense- and RNA interference-based approaches have been identified. These provide targeted approaches to delivery of antinociceptive molecules or interruption of pain pathways without subjecting the patient to systemic toxicity of drugs. Some of these approaches are aimed at correcting the underlying pathology of the diseases (e.g., treating degenerative joint diseases causing pain). Management of neuropathic pain is a challenge and a number of studies are addressing it. Overall the future of gene therapy for pain is promising.
Collapse
Affiliation(s)
- K K Jain
- Jain PharmaBiotech, Blaesiring 7, CH-4057, Basel, Switzerland.
| |
Collapse
|
74
|
Abstract
The prevalence of people suffering from chronic pain is extremely high and pain affects millions of people worldwide. As such, persistent pain represents a major health problem and an unmet clinical need. The reason for the high incidence of chronic pain patients is in a large part due to a paucity of effective pain control. An important reason for poor pain control is undoubtedly a deficit in our understanding of the underlying causes of chronic pain and as a consequence our arsenal of analgesic therapies is limited. However, there is considerable hope for the development of new classes of analgesic drugs by targeting novel processes contributing to clinically relevant pain. In this chapter we highlight a number of molecular species which are potential therapeutic targets for future neuropathic pain treatments. In particular, the roles of voltage-gated ion channels, neuroinflammation, protein kinases and neurotrophins are discussed in relation to the generation of neuropathic pain and how by targeting these molecules it may be possible to provide better pain control than is currently available.
Collapse
Affiliation(s)
- Fabien Marchand
- King's College London, London, Neurorestoration, CARD Wolfson Wing, Hodgkin Building, Guy's Campus, London Bridge, London, SE1 1UL, UK
| | | | | |
Collapse
|
75
|
Glorioso JC, Fink DJ. Herpes vector-mediated gene transfer in the treatment of chronic pain. Mol Ther 2008; 17:13-8. [PMID: 18841093 DOI: 10.1038/mt.2008.213] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Chronic pain is a major health concern with up to 50% of patients finding little if any relief following traditional pharmacotherapy. This review describes the treatment of chronic pain using herpes simplex virus type 1 (HSV)-based vectors. HSV can be effectively used to deliver pain-modulating transgenes to sensory neurons in vivo following intradermal inoculation. The vector genome persists in peripheral nerve bodies in an episomal state and serves as a platform for expression of natural pain-relieving molecules that access endogenous antinociceptive circuitry. The vectors are mutated to prevent reactivation from latency or spread to the central nervous system. Dermatome selection for administration of HSV vectors provides targeted delivery of pain gene therapy to primary afferent neurons. This novel approach alleviates pain without systemic side effects or the induction of tolerance and can be used in combination with standard pain treatments.
Collapse
Affiliation(s)
- Joseph C Glorioso
- 1Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
76
|
Abstract
PURPOSE OF REVIEW Recent studies show that peripheral injury activates both neuronal and nonneuronal or glial components of the peripheral and central cellular circuitry. The subsequent neuron-glia interactions contribute to pain hypersensitivity. This review will briefly discuss novel findings that have shed light on the cellular mechanisms of neuron-glia interactions in persistent pain. RECENT FINDINGS Two fundamental questions related to neuron-glia interactions in pain mechanisms have been addressed: what are the signals that lead to central glial activation after injury and how do glial cells affect central nervous system neuronal activity and promote hyperalgesia? SUMMARY Evidence indicates that central glial activation depends on nerve inputs from the site of injury and release of chemical mediators. Hematogenous immune cells may migrate to/infiltrate the brain and circulating inflammatory mediators may penetrate the blood-brain barrier to participate in central glial responses to injury. Inflammatory cytokines such as interleukin-1beta released from glia may facilitate pain transmission through its coupling to neuronal glutamate receptors. This bidirectional neuron-glia signaling plays a key role in glial activation, cytokine production and the initiation and maintenance of hyperalgesia. Recognition of the contribution of the mutual neuron-glia interactions to central sensitization and hyperalgesia prompts new treatment for chronic pain.
Collapse
Affiliation(s)
- Ke Ren
- Department of Neural and Pain Sciences, Dental School and Program in Neuroscience, University of Maryland, Baltimore, Maryland 21201-1586, USA
| | | |
Collapse
|
77
|
Mata M, Hao S, Fink DJ. Gene therapy directed at the neuroimmune component of chronic pain with particular attention to the role of TNF alpha. Neurosci Lett 2008; 437:209-13. [PMID: 18403116 PMCID: PMC2668118 DOI: 10.1016/j.neulet.2008.03.049] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Revised: 02/28/2008] [Accepted: 03/19/2008] [Indexed: 01/17/2023]
Abstract
Identification that neuroimmune activation in the spinal cord is an important factor in the development of chronic pain has opened the possibility that gene transfer of anti-inflammatory peptides may be used to reduce pain neurotransmission. We review the published evidence regarding gene transfer to meninges to express the anti-inflammatory peptide interleukin 10, and gene transfer to dorsal root ganglia using replication incompetent HSV vectors to express interleukin 4, interleukin 10, or the soluble (p55) tumor necrosis factor receptor (sTNFR). The results of these experiments suggest a novel role for "reverse signaling" through the full-length membrane form of TNFalpha in spinal glia in the modulation of chronic pain.
Collapse
Affiliation(s)
- Marina Mata
- Department of Neurology, University of Michigan, 1914 Taubman Center, 1500 E. Medical Center Drive, Ann Arbor, MI 48109-0316, USA; VA Ann Arbor Healthcare System, Ann Arbor, MI 48109-0316, USA
| | | | | |
Collapse
|
78
|
Abstract
Chronic pain is a highly prevalent condition that impacts adversely on individual quality of life, imposes substantial costs on the healthcare system and a considerable burden on society. Advances in the understanding of pain mechanisms have opened the way for the development of new treatment strategies. The continuous delivery of short-lived potent bioactive molecules to sensory nerves, spinal cord or meninges--achieved by directed gene transfer--offers the possibility to selectively interrupt nociceptive neurotransmission or to interfere with the plastic changes in the nervous system underlying the development or persistence of chronic pain. In this review we describe advances in the use of non-viral and viral vector-based gene transfer for the treatment of pain, with a special focus on the use of recombinant non-replicating herpes simplex virus-based vectors and the prospects for clinical trials.
Collapse
Affiliation(s)
- Marina Mata
- Department of Neurology, University of Michigan School of Medicine and VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | | | | |
Collapse
|
79
|
Smith SF, Blackman G, Hopper C. Numb chin syndrome: a nonmetastatic neurological manifestation of malignancy. ACTA ACUST UNITED AC 2008; 105:e53-6. [PMID: 18280947 DOI: 10.1016/j.tripleo.2007.11.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Revised: 11/11/2007] [Accepted: 11/13/2007] [Indexed: 12/21/2022]
Abstract
Numb chin syndrome, sometimes called numb lip syndrome, is an uncommon but well-recognized symptom in medical oncology. It may be a nonmetastatic neurological manifestation of malignancy, often with no clinically visible pathology. We report a case of a 79-year-old woman with widespread metastatic disease secondary to breast carcinoma who presented with a left-sided numb lip and chin. The patient had no clinical or radiographic pathology within the head and neck region. Numb chin syndrome is almost unknown within the dental and oral and maxillofacial community, despite being well reported in the medical literature. We urge all general dentists, oral medicine specialists, and oral and maxillofacial surgeons to consider metastatic cancer in patients with unexplained facial hypoesthesia.
Collapse
Affiliation(s)
- Samantha Fleur Smith
- Department of Oral and Maxillofacial Surgery, University College London Hospital, London, United Kingdom.
| | | | | |
Collapse
|
80
|
Abstract
Cytokine activation or dysregulation is implied in a variety of painful disease states. Numerous experimental studies provide evidence that proinflammatory cytokines induce or facilitate neuropathic pain. Cytokine levels are rapidly and markedly upregulated in the peripheral nerves, dorsal root ganglia, spinal cord and in particular regions of the brain, after peripheral nerve injuries. Direct receptor-mediated actions on afferent nerve fibers as well as cytokine effects involving further mediators have been reported. Whereas direct application of exogenous proinflammatory cytokines induces pain, blockade of these cytokines or application of anti-inflammatory cytokines reduces pain behavior in most experimental paradigms. Cytokine measurements may identify patients at risk of developing chronic pain associated with their neuropathic conditions, as in the examples of peripheral neuropathies and postherpetic neuralgia. Anticytokine agents currently on the market are effective for the treatment of mostly inflammatory pain conditions, and are starting to be introduced for neuropathic pain states; however, their use is limited by potential life-threatening complications. Owing to the pleiotropy and redundancy of the cytokine system, the successful approach may not be inhibition of one particular cytokine but strategies shifting the balance between pro- and anti-inflammatory cytokines in properly selected patients. Agents that specifically target downstream signaling molecules may provide hope for safer and more specific therapies.
Collapse
Affiliation(s)
- Maria Schäfers
- Department of Neurology, University of Duisburg-Essen, Hufelandstr. 55,45147 Essen, Germany.
| | | |
Collapse
|