1
|
Ingusci S, Goins WF, Cohen JB, Miyagawa Y, Knipe DM, Glorioso JC. Next-generation replication-defective HSV vectors for delivery of large DNA payloads. Mol Ther 2025:S1525-0016(25)00264-3. [PMID: 40181547 DOI: 10.1016/j.ymthe.2025.03.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025] Open
Abstract
The application of gene therapy to the treatment of human disease with complex etiology and pathology will often require the delivery of large payloads exceeding 10 kbp in size. This is generally not possible with the most popular vectors such as adeno-associated viruses (AAVs), lentiviruses (LVs), retroviruses (RVs), and many nonviral delivery systems. There is a high likelihood that the correction of many human gene defects such as those associated with neurodegenerative diseases and inflammatory processes will require single large genes or complex genetic payloads that will often necessitate precise regulatory control of the specificity, timing, and duration of corrective gene expression. The regulation of cellular gene products typically depends on genomic promoter systems and splicing-driven transcription variants, necessitating a delivery vector with substantial payload capacity. Replication-defective herpes simplex virus (rdHSV) mutants lack at least one essential viral gene product and are propagated in host cells that supply the missing gene product. This review explores next-generation rdHSV vectors, which do not express viral genes, offer high payload capacity, and can be engineered for safe, long-term transgene expression. These advanced vectors enable the correction of complex diseases affecting neurons and other tissues, paving the way for large or intricate gene replacement strategies.
Collapse
Affiliation(s)
- Selene Ingusci
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - William F Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Justus B Cohen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Yoshitaka Miyagawa
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo 113-8602, Japan
| | - David M Knipe
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
2
|
da Silva A, Lepetre-Mouelhi S, Couvreur P. Micro- and nanocarriers for pain alleviation. Adv Drug Deliv Rev 2022; 187:114359. [PMID: 35654211 DOI: 10.1016/j.addr.2022.114359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 12/28/2022]
Abstract
Acute or chronic pain is a major source of impairment in quality of life and affects a substantial part of the population. To date, pain is alleviated by a limited range of treatments with significant toxicity, increased risk of misuse and inconsistent efficacy, owing, in part, to lack of specificity and/or unfavorable pharmacokinetic properties. Thanks to the unique properties of nanoscaled drug carriers, nanomedicine may enhance drug biodistribution and targeting, thus contributing to improved bioavailability and lower off-target toxicity. After a brief overview of the current situation and the main critical issues regarding pain alleviation, this review will examine the most advanced approaches using nanomedicine of each drug class, from the preclinical stage to approved nanomedicines.
Collapse
|
3
|
Wang T, Zhu X, Yi H, Gu J, Liu S, Izenwasser S, Lemmon VP, Roy S, Hao S. Viral vector-mediated gene therapy for opioid use disorders. Exp Neurol 2021; 341:113710. [PMID: 33781732 DOI: 10.1016/j.expneurol.2021.113710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/26/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022]
Abstract
Chronic exposure to opioids typically results in adverse consequences. Opioid use disorder (OUD) is a disease of the CNS with behavioral, psychological, neurobiological, and medical manifestations. OUD induces a variety of changes of neurotransmitters/neuropeptides in the nervous system. Existing pharmacotherapy, such as opioid maintenance therapy (OMT) is the mainstay for the treatment of OUD, however, current opioid replacement therapy is far from effective for the majority of patients. Pharmacological therapy for OUD has been challenging for many reasons including debilitating side-effects. Therefore, developing an effective, non-pharmacological approach would be a critical advancement in improving and expanding treatment for OUD. Viral vector mediated gene therapy provides a potential new approach for treating opioid abused patients. Gene therapy can supply targeting gene products directly linked to the mechanisms of OUD to restore neurotransmitter and/or neuropeptides imbalance, and avoid the off-target effects of systemic administration of drugs. The most commonly used viral vectors in rodent studies of treatment of opioid-used disorder are based on recombinant adenovirus (AV), adeno-associated virus (AAV), lentiviral (LV) vectors, and herpes simplex virus (HSV) vectors. In this review, we will focus on the recent progress of viral vector mediated gene therapy in OUD, especially morphine tolerance and withdrawal.
Collapse
Affiliation(s)
- Tao Wang
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Xun Zhu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Hyun Yi
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Jun Gu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shue Liu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sari Izenwasser
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Vance P Lemmon
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shuanglin Hao
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America.
| |
Collapse
|
4
|
Godai K, Takahashi K, Kashiwagi Y, Liu CH, Yi H, Liu S, Dong C, Lubarsky DA, Hao S. Ryanodine Receptor to Mitochondrial Reactive Oxygen Species Pathway Plays an Important Role in Chronic Human Immunodeficiency Virus gp120MN-Induced Neuropathic Pain in Rats. Anesth Analg 2020; 129:276-286. [PMID: 30507840 DOI: 10.1213/ane.0000000000003916] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Chronic pain is one of the most common complaints in patients with human immunodeficiency virus (HIV)-associated sensory neuropathy. Ryanodine receptor (RyR) and mitochondrial oxidative stress are involved in neuropathic pain induced by nerve injury. Here, we investigated the role of RyR and mitochondrial superoxide in neuropathic pain induced by repeated intrathecal HIV glycoprotein 120 (gp120) injection. METHODS Recombinant HIV glycoprotein gp120MN was intrathecally administered to induce neuropathic pain. Mechanical threshold was tested using von Frey filaments. Peripheral nerve fiber was assessed by the quantification of the intraepidermal nerve fiber density in the skin of the hindpaw. The expression of spinal RyR was examined using Western blots. Colocalization of RyR with neuronal nuclei (NeuN; neuron marker), glial fibrillary acidic protein (GFAP; astrocyte marker), or ionizing calcium-binding adaptor molecule 1 (Iba1; microglia marker) in the spinal cord was examined using immunohistochemistry. MitoSox-positive profiles (a mitochondrial-targeted fluorescent superoxide indicator) were examined. The antiallodynic effects of intrathecal administration of RyR antagonist, dantrolene (a clinical drug for malignant hyperthermia management), or selective mitochondrial superoxide scavenger, Mito-Tempol, were evaluated in the model. RESULTS We found that repeated but not single intrathecal injection of recombinant protein gp120 induced persistent mechanical allodynia. Intraepidermal nerve fibers in repeated gp120 group was lower than that in sham at 2 weeks, and the difference in means (95% confidence interval) was 8.495 (4.79-12.20), P = .0014. Repeated gp120 increased expression of RyR, and the difference in means (95% confidence interval) was 1.50 (0.504-2.495), P = .007. Repeated gp120 also increased mitochondrial superoxide cell number in the spinal cord, and the difference in means (95% confidence interval) was 6.99 (5.99-8.00), P < .0001. Inhibition of spinal RyR or selective mitochondrial superoxide scavenger dose dependently reduced mechanical allodynia induced by repeated gp120 injection. RyR and mitochondrial superoxide were colocalized in the neuron, but not glia. Intrathecal injection of RyR inhibitor lowered mitochondrial superoxide in the spinal cord dorsal horn in the gp120 neuropathic pain model. CONCLUSIONS These data suggest that repeated intrathecal HIV gp120 injection induced an acute to chronic pain translation in rats, and that neuronal RyR and mitochondrial superoxide in the spinal cord dorsal horn played an important role in the HIV neuropathic pain model. The current results provide evidence for a novel approach to understanding the molecular mechanisms of HIV chronic pain and treating chronic pain in patients with HIV.
Collapse
Affiliation(s)
| | | | | | | | - Hyun Yi
- From the Departments of Anesthesiology
| | - Shue Liu
- From the Departments of Anesthesiology
| | - Chuanhui Dong
- Neurology, University of Miami Miller School of Medicine, Miami, Florida
| | | | | |
Collapse
|
5
|
Zhang T, Zhang N, Zhang R, Zhao W, Chen Y, Wang Z, Xu B, Zhang M, Shi X, Zhang Q, Guo Y, Xiao J, Chen D, Fang Q. Preemptive intrathecal administration of endomorphins relieves inflammatory pain in male mice via inhibition of p38 MAPK signaling and regulation of inflammatory cytokines. J Neuroinflammation 2018; 15:320. [PMID: 30442166 PMCID: PMC6236886 DOI: 10.1186/s12974-018-1358-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/02/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Preemptive administration of analgesic drugs reduces perceived pain and prolongs duration of antinociceptive action. Whereas several lines of evidence suggest that endomorphins, the endogenous mu-opioid agonists, attenuate acute and chronic pain at the spinal level, their preemptive analgesic effects remain to be determined. In this study, we evaluated the anti-allodynic activities of endomorphins and explored their mechanisms of action after preemptive administration in a mouse model of inflammatory pain. METHODS The anti-allodynic activities of preemptive intrathecal administration of endomorphin-1 and endomorphin-2 were investigated in complete Freund's adjuvant (CFA)-induced inflammatory pain model and paw incision-induced postoperative pain model. The modulating effects of endomorphins on the expression of p38 mitogen-activated protein kinase (p38 MAPK) and inflammatory mediators in dorsal root ganglion (DRG) of CFA-treated mice were assayed by real-time reverse transcription-polymerase chain reaction (RT-PCR), Western blotting, or immunofluorescence staining. RESULTS Preemptive intrathecal injection of endomorphins dose-dependently attenuated CFA-induced mechanical allodynia via the mu-opioid receptor and significantly reversed paw incision-induced allodynia. In addition, CFA-caused increase of phosphorylated p38 MAPK in DRG was dramatically reduced by preemptive administration of endomorphins. Repeated intrathecal application of the specific p38 MAPK inhibitor SB203580 reduced CFA-induced mechanical allodynia as well. Further RT-PCR assay showed that endomorphins regulated the mRNA expression of inflammatory cytokines in DRGs induced by peripheral inflammation. CONCLUSIONS Our findings reveal a novel mechanism by which preemptive treatment of endomorphins attenuates inflammatory pain through regulating the production of inflammatory cytokines in DRG neurons via inhibition of p38 MAPK phosphorylation.
Collapse
Affiliation(s)
- Ting Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Nan Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Run Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Weidong Zhao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Yong Chen
- Department of Neurology, School of Medicine, Duke University, Durham, North Carolina, 27710, USA
| | - Zilong Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Biao Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Mengna Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Xuerui Shi
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Qinqin Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Yuanyuan Guo
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Jian Xiao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Dan Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Quan Fang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
6
|
Abstract
Virus-mediated gene delivery shows promise for the treatment of chronic pain. However, viral vectors have cytotoxicity. To avoid toxicities and limitations of virus-mediated gene delivery, we developed a novel nonviral hybrid vector: HIV-1 Tat peptide sequence modified with histidine and cysteine residues combined with a cationic lipid. The vector has high transfection efficiency with little cytotoxicity in cancer cell lines including HSC-3 (human tongue squamous cell carcinoma) and exhibits differential expression in HSC-3 (∼45-fold) relative to HGF-1 (human gingival fibroblasts) cells. We used the nonviral vector to transfect cancer with OPRM1, the μ-opioid receptor gene, as a novel method for treating cancer-induced pain. After HSC-3 cells were transfected with OPRM1, a cancer mouse model was created by inoculating the transfected HSC-3 cells into the hind paw or tongue of athymic mice to determine the analgesic potential of OPRM1 transfection. Mice with HSC-3 tumors expressing OPRM1 demonstrated significant antinociception compared with control mice. The effect was reversible with local naloxone administration. We quantified β-endorphin secretion from HSC-3 cells and showed that HSC-3 cells transfected with OPRM1 secreted significantly more β-endorphin than control HSC-3 cells. These findings indicate that nonviral delivery of the OPRM1 gene targeted to the cancer microenvironment has an analgesic effect in a preclinical cancer model, and nonviral gene delivery is a potential treatment for cancer pain.
Collapse
|
7
|
Hu C, Cai Z, Lu Y, Cheng X, Wu Z, Zhang Q. Local intramuscular injection of a plasmid encoding human proenkepahlin attenuates incision pain in rats. Neurosci Lett 2016; 632:157-62. [DOI: 10.1016/j.neulet.2016.08.058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 08/25/2016] [Accepted: 08/30/2016] [Indexed: 11/25/2022]
|
8
|
Hu C, Cai Z, Lu Y, Cheng X, Guo Q, Wu Z, Zhang Q. Nonviral vector plasmid DNA encoding human proenkephalin gene attenuates inflammatory and neuropathic pain-related behaviors in mice. Neurosci Lett 2016; 634:87-93. [PMID: 27693568 DOI: 10.1016/j.neulet.2016.09.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/17/2016] [Accepted: 09/22/2016] [Indexed: 12/21/2022]
Abstract
Inflammatory pain and neuropathic pain are major clinical health issues that represent considerable social and economic burden worldwide. In the present study, we investigated the anti-nociceptive efficacy of delivery of human proenkephalin gene by a plasmid DNA vector (pVAX1-PENK) on complete Freund's adjuvant (CFA) induced inflammatory pain and spared nerve injury (SNI) induced neuropathic pain in mice. Mice were intramuscularly or intrathecally administered pVAX1 or pVAX1-PENK, respectively. Pain thresholds in the pVAX1-PENK treated mice were significantly higher at day 3, then reached a peak at day 7 and lasted until day 28 after gene transfer, and the analgesic effect of pVAX1-PENK was blocked with naloxone hydrochloride. In contrast, pVAX1 treated mice did not significantly improve pain thresholds. These results indicate that peripheral or spinal delivery of a plasmid encoding human proenkephalin gene provides a potential therapeutic strategy for inflammatory pain and neuropathic pain.
Collapse
Affiliation(s)
- Chunsheng Hu
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China; College of Life Science and Bioengineering, Beijing University of Technology, Beijing 10024, People's Republic of China
| | - Zhenzhen Cai
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China
| | - Yuxin Lu
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China
| | - Xiaochen Cheng
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China
| | - Qi Guo
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China
| | - Zuze Wu
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China; College of Life Science and Bioengineering, Beijing University of Technology, Beijing 10024, People's Republic of China
| | - Qinglin Zhang
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China.
| |
Collapse
|
9
|
Kanda H, Liu S, Iida T, Yi H, Huang W, Levitt RC, Lubarsky DA, Candiotti KA, Hao S. Inhibition of Mitochondrial Fission Protein Reduced Mechanical Allodynia and Suppressed Spinal Mitochondrial Superoxide Induced by Perineural Human Immunodeficiency Virus gp120 in Rats. Anesth Analg 2016; 122:264-72. [PMID: 26418124 DOI: 10.1213/ane.0000000000000962] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Mitochondria play an important role in many cellular and physiologic functions. Mitochondria are dynamic organelles, and their fusion and fission regulate cellular signaling, development, and mitochondrial homeostasis. The most common complaint of human immunodeficiency virus (HIV)-sensory neuropathy is pain on the soles in patients with HIV, but the exact molecular mechanisms of HIV neuropathic pain are not clear. In the present study, we investigated the role of mitochondrial dynamin-related protein 1 (Drp1, a GTPase that mediates mitochondrial fission) in the perineural HIV coat glycoprotein gp120-induced neuropathic pain state. METHODS Neuropathic pain was induced by the application of recombinant HIV-1 envelope protein gp120 into the sciatic nerve. Mechanical threshold was tested using von Frey filaments. The mechanical threshold response was assessed over time using the area under curves. Intrathecal administration of antisense oligodeoxynucleotide (ODN) against Drp1, mitochondrial division inhibitor-1 (mdivi-1), or phenyl-N-tert-butylnitrone (a reactive oxygen species scavenger) was given. The expression of spinal Drp1 was examined using western blots. The expression of mitochondrial superoxide in the spinal dorsal horn was examined using MitoSox imaging. RESULTS Intrathecal administration of either antisense ODN against Drp1 or mdivi-1 decreased mechanical allodynia (a sensation of pain evoked by nonpainful stimuli) in the gp120 model. Intrathecal ODN or mdivi-1 did not change basic mechanical threshold in sham surgery rats. Intrathecal Drp1 antisense ODN decreased the spinal expression of increased Drp1 protein induced by peripheral gp120 application. Intrathecal phenyl-N-tert-butylnitrone reduced mechanical allodynia. Furthermore, both intrathecal Drp1 antisense ODN and mdivi-1 reversed the upregulation of mitochondrial superoxide in the spinal dorsal horn in the gp120 neuropathic pain state. CONCLUSIONS These data suggest that mitochondrial division plays a substantial role in the HIV gp120-related neuropathic pain state through mitochondrial reactive oxygen species and provides evidence for a novel approach to treating chronic pain in patients with HIV.
Collapse
Affiliation(s)
- Hirotsugu Kanda
- From the *Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida; †Department of Anesthesiology, Asahikawa Medical University, Asahikawa, Japan; ‡Hussman Institute of Human Genomics, University of Miami Miller School of Medicine, Miami, Florida; and §Veterans Affairs Medical Center, Miami, Florida
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Kanda H, Kanao M, Liu S, Yi H, Iida T, Levitt RC, Candiotti KA, Lubarsky DA, Hao S. HSV vector-mediated GAD67 suppresses neuropathic pain induced by perineural HIV gp120 in rats through inhibition of ROS and Wnt5a. Gene Ther 2016; 23:340-8. [PMID: 26752351 PMCID: PMC4824655 DOI: 10.1038/gt.2016.3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 11/22/2015] [Accepted: 12/31/2015] [Indexed: 12/19/2022]
Abstract
Human immunodeficiency virus (HIV)-related neuropathic pain is a debilitating chronic condition that is severe and unrelenting. Despite the extensive research, the exact neuropathological mechanisms remain unknown, which hinders our ability to develop effective treatments. Loss of GABAergic tone may play an important role in the neuropathic pain state. Glutamic acid decarboxylase 67 (GAD67) is one of isoforms that catalyze GABA synthesis. Here, we used recombinant herpes simplex virus (HSV-1) vectors that encode gad1 gene to evaluate the therapeutic potential of GAD67 in peripheral HIV gp120-induced neuropathic pain in rats. We found that 1) subcutaneous inoculation of the HSV vectors expressing GAD67 attenuated mechanical allodynia in the model of HIV gp120-induced neuropathic pain, 2) the anti-allodynic effect of GAD67 was reduced by GABA-A and-B receptors antagonists, 3) HSV vectors expressing GAD67 reversed the lowered GABA-IR expression, and 4) the HSV vectors expressing GAD67 suppressed the upregulated mitochondrial superoxide and Wnt5a in the spinal dorsal horn. Taken together, our studies support the concept that recovering GABAergic tone by the HSV vectors may reverse HIV-associated neuropathic pain through suppressing mitochondrial superoxide and Wnt5a. Our studies provide validation of HSV-mediated GAD67 gene therapy in the treatment of HIV-related neuropathic pain.
Collapse
Affiliation(s)
- H Kanda
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Anesthesiology, Asahikawa Medical University, Asahikawa, Japan
| | - M Kanao
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Anesthesiology, Asahikawa Medical University, Asahikawa, Japan
| | - S Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - H Yi
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - T Iida
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Anesthesiology, Asahikawa Medical University, Asahikawa, Japan
| | - R C Levitt
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, USA.,Hussman Institute of Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA.,Veterans Affairs Medical Center, Miami, FL, USA
| | - K A Candiotti
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - D A Lubarsky
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - S Hao
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
11
|
Wolfe D, Krisky D, Goss J, Wechuck J, Mata M, Fink DJ. Translating Gene Therapy for Pain from Animal Studies to the Clinic. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
12
|
Kibaly C, Loh H, Law PY. A Mechanistic Approach to the Development of Gene Therapy for Chronic Pain. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 327:89-161. [DOI: 10.1016/bs.ircmb.2016.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
13
|
Kanao M, Kanda H, Huang W, Liu S, Yi H, Candiotti KA, Lubarsky DA, Levitt RC, Hao S. Gene Transfer of Glutamic Acid Decarboxylase 67 by Herpes Simplex Virus Vectors Suppresses Neuropathic Pain Induced by Human Immunodeficiency Virus gp120 Combined with ddC in Rats. Anesth Analg 2015; 120:1394-404. [PMID: 25851180 DOI: 10.1213/ane.0000000000000729] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Human immunodeficiency virus (HIV)-related painful sensory neuropathies primarily consist of the HIV infection-related distal sensory polyneuropathy and antiretroviral toxic neuropathies. Pharmacotherapy provides only partial relief of pain in patients with HIV/acquired immune deficiency syndrome because little is known about the exact neuropathological mechanisms for HIV-associated neuropathic pain (NP). Hypofunction of γ-aminobutyric acid (GABA) GABAergic inhibitory mechanisms has been reported after peripheral nerve injury. In this study, we tested the hypothesis that HIV gp120 combined with antiretroviral therapy reduces spinal GABAergic inhibitory tone and that restoration of GABAergic inhibitory tone will reduce HIV-related NP in a rat model. METHODS The application of recombinant HIV-1 envelope protein gp120 into the sciatic nerve plus systemic ddC (one antiretroviral drug) induced mechanical allodynia. The hind paws of rats were inoculated with replication-defective herpes simplex virus (HSV) vectors genetically encoding gad1 gene to express glutamic acid decarboxylase 67 (GAD67), an enzyme that catalyzes the decarboxylation of glutamate to GABA. Mechanical threshold was tested using von Frey filaments before and after treatments with the vectors. The expression of GAD67 in both the lumbar spinal cord and the L4-5 dorsal root ganglia was examined using western blots. The expression of mitochondrial superoxide in the spinal dorsal horn was examined using MitoSox imaging. The immunoreactivity of spinal GABA, pCREB, and pC/EBPβ was tested using immunohistochemistry. RESULTS In the gp120 with ddC-induced neuropathic pain model, GAD67 expression mediated by the HSV vector caused an elevation of mechanical threshold that was apparent on day 3 after vector inoculation. The antiallodynic effect of the single HSV vector inoculation expressing GAD67 lasted >28 days. The area under the time-effect curves in the HSV vector expressing GAD67 was increased compared with that in the control vectors (P = 0.0005). Intrathecal GABA-A/B agonists elevated mechanical threshold in the pain model. The HSV vectors expressing GAD67 reversed the lowered GABA immunoreactivity in the spinal dorsal horn in the neuropathic rats. HSV vectors expressing GAD67 in the neuropathic rats reversed the increased signals of mitochondrial superoxide in the spinal dorsal horn. The vectors expressing GAD67 reversed the upregulated immunoreactivity expression of pCREB and pC/EBPβ in the spinal dorsal horn in rats exhibiting NP. CONCLUSIONS Based on our results, we suggest that GAD67 mediated by HSV vectors acting through the suppression of mitochondrial reactive oxygen species and transcriptional factors in the spinal cord decreases pain in the HIV-related neuropathic pain model, providing preclinical evidence for gene therapy applications in patients with HIV-related pain states.
Collapse
Affiliation(s)
- Megumi Kanao
- From the *Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida; †Department of Anesthesiology, Asahikawa Medical University, Asahikawa, Japan; ‡Hussman Institute of Human Genomics, University of Miami Miller School of Medicine, Miami, Florida; and §Veterans Affairs Medical Center, Miami, Florida
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Guedon JMG, Wu S, Zheng X, Churchill CC, Glorioso JC, Liu CH, Liu S, Vulchanova L, Bekker A, Tao YX, Kinchington PR, Goins WF, Fairbanks CA, Hao S. Current gene therapy using viral vectors for chronic pain. Mol Pain 2015; 11:27. [PMID: 25962909 PMCID: PMC4446851 DOI: 10.1186/s12990-015-0018-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/01/2015] [Indexed: 02/07/2023] Open
Abstract
The complexity of chronic pain and the challenges of pharmacotherapy highlight the importance of development of new approaches to pain management. Gene therapy approaches may be complementary to pharmacotherapy for several advantages. Gene therapy strategies may target specific chronic pain mechanisms in a tissue-specific manner. The present collection of articles features distinct gene therapy approaches targeting specific mechanisms identified as important in the specific pain conditions. Dr. Fairbanks group describes commonly used gene therapeutics (herpes simplex viral vector (HSV) and adeno-associated viral vector (AAV)), and addresses biodistribution and potential neurotoxicity in pre-clinical models of vector delivery. Dr. Tao group addresses that downregulation of a voltage-gated potassium channel (Kv1.2) contributes to the maintenance of neuropathic pain. Alleviation of chronic pain through restoring Kv1.2 expression in sensory neurons is presented in this review. Drs Goins and Kinchington group describes a strategy to use the replication defective HSV vector to deliver two different gene products (enkephalin and TNF soluble receptor) for the treatment of post-herpetic neuralgia. Dr. Hao group addresses the observation that the pro-inflammatory cytokines are an important shared mechanism underlying both neuropathic pain and the development of opioid analgesic tolerance and withdrawal. The use of gene therapy strategies to enhance expression of the anti-pro-inflammatory cytokines is summarized. Development of multiple gene therapy strategies may have the benefit of targeting specific pathologies associated with distinct chronic pain conditions (by Guest Editors, Drs. C. Fairbanks and S. Hao).
Collapse
Affiliation(s)
- Jean-Marc G Guedon
- Graduate Program in Molecular Virology and Microbiology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA. .,Department of Ophthalmology, University of Pittsburgh School of Medicine, Room 1020 EEI, 203 Lothrop Street, Pittsburgh, PA, 15213, USA.
| | - Shaogen Wu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, State University of New Jersey, 185 S. Orange Ave., MSB, F-548, Newark, NJ, 07103, USA.
| | - Xuexing Zheng
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | | | - Joseph C Glorioso
- Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 424 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
| | - Ching-Hang Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Shue Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.
| | - Alex Bekker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, State University of New Jersey, 185 S. Orange Ave., MSB, F-548, Newark, NJ, 07103, USA.
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, State University of New Jersey, 185 S. Orange Ave., MSB, F-548, Newark, NJ, 07103, USA. .,Department of Cell Biology & Molecular Medicine, New Jersey Medical School, Rutgers, State University of New Jersey, Newark, NJ, 07103, USA. .,Department of Neurology & Neuroscience, New Jersey Medical School, Rutgers, State University of New Jersey, Newark, NJ, 07103, USA. .,Department of Physiology & Pharmacology, New Jersey Medical School, Rutgers, State University of New Jersey, Newark, NJ, 07103, USA.
| | - Paul R Kinchington
- Graduate Program in Molecular Virology and Microbiology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA. .,Department of Ophthalmology, University of Pittsburgh School of Medicine, Room 1020 EEI, 203 Lothrop Street, Pittsburgh, PA, 15213, USA.
| | - William F Goins
- Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 424 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
| | - Carolyn A Fairbanks
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA. .,Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA. .,Department of Pharmacology, University of Minnesota, 9-177 Weaver Densford Hall, 308 Harvard Street, Minneapolis, MN, 55455, USA.
| | - Shuanglin Hao
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
15
|
Wu XN, Zhang T, Qian NS, Guo XD, Yang HJ, Huang KB, Luo GQ, Xiang W, Deng WT, Dai GH, Peng KR, Pan SY. Antinociceptive effects of endomorphin-2: suppression of substance P release in the inflammatory pain model rat. Neurochem Int 2015; 82:1-9. [PMID: 25661513 DOI: 10.1016/j.neuint.2015.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 01/02/2015] [Accepted: 01/13/2015] [Indexed: 12/13/2022]
Abstract
Endomorphin-2 (EM2) and Substance P (SP) exert suppressive and facilitative influences upon nociception, respectively. Although EM2 and SP were often co-expressed in single neurons in dorsal root ganglion (DRG), it is still unknown if and how the nociception-suppressive influences of EM2 might be exerted upon nociception-facilitative effects of SP in the DRG neurons. We examined these issues in the inflammatory pain model rats produced by subcutaneous injection of the complete Freund's adjuvant into the hind paw. The paw withdrawal threshold for mechanical allodynia was measured. Changes of EM2 and SP release were estimated by measuring intrathecal levels of EM2 and SP through in vivo microdialysis analysis of cerebrospinal fluid. The mechanical allodynia was dose-dependently attenuated by intrathecal injection of EM2 or a neurokinin-1 receptor antagonist, and facilitated by intrathecal injection of SP or a mu-opioid receptor (MOR) antagonist. Importantly, intrathecal level of SP was found to be lowered by intrathecal injection of EM2. Morphologically, colocalization of EM2-, MOR- and SP-immunoreactivity in single DRG neurons was observed by immunofluorescent histochemistry, and co-expression of EM2 and SP in large, dense-cored presynaptic vesicles in primary afferents, as well as localization of MOR on pre- and postsynaptic membrane in spinal dorsal horn, was also confirmed electron miscroscopically. Thus, the results indicated that analgesic influences of EM2 upon inflammatory pain might be exerted through suppression of SP release, supporting the assumptions that binding of EM2 to presynaptic MOR might induce such effects.
Collapse
Affiliation(s)
- Xiao-Na Wu
- Department of Neurology, Nanfang Hospital of Southern Medical University, Guangzhou 510515, China; Department of Neurology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - Tao Zhang
- Department of Orthopaedics, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - Nian-Song Qian
- Department of Oncology Unit 2, PLA General Hospital, Beijing 100853, China
| | - Xiao-Dong Guo
- Department of Neurosurgery, The 153th Hospital of PLA, Zhengzhou 450042, China
| | - Hong-Jun Yang
- Department of Neurology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - Kai-Bin Huang
- Department of Neurology, Nanfang Hospital of Southern Medical University, Guangzhou 510515, China
| | - Gao-Quan Luo
- Department of Neurology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - Wei Xiang
- Department of Neurology, Nanfang Hospital of Southern Medical University, Guangzhou 510515, China; Department of Neurology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - Wen-Ting Deng
- Department of Neurology, Nanfang Hospital of Southern Medical University, Guangzhou 510515, China
| | - Guang-Hai Dai
- Department of Oncology Unit 2, PLA General Hospital, Beijing 100853, China.
| | - Kai-Run Peng
- Department of Neurology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, China.
| | - Su-Yue Pan
- Department of Neurology, Nanfang Hospital of Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
16
|
Injury-specific promoters enhance herpes simplex virus-mediated gene therapy for treating neuropathic pain in rodents. THE JOURNAL OF PAIN 2015; 16:283-90. [PMID: 25576797 DOI: 10.1016/j.jpain.2014.12.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 12/13/2014] [Accepted: 12/13/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED Chronic neuropathic pain is often difficult to treat with current pain medications. Gene therapy is presently being explored as a therapeutic approach for the treatment of neuropathic and cancer pain. In this study, we sought to use an injury-specific promoter to deliver the mu-opioid receptor (MOR) transgene such that expression would occur during the injured state only in response to release of injury-specific galanin. To determine whether an injury-specific promoter can produce neuron-specific MOR expression and enhanced antinociception, we compared animals infected with a galanin promoter virus (galMOR) or a human cytomegalovirus promoter virus (cmvMOR). In behavioral assays, we found an earlier onset and a larger magnitude of antinociception in animals infected with galMOR compared with cmvMOR. Immunohistochemical analysis of dorsal root ganglion neurons revealed a significant increase in MOR-positive staining in cmvMOR- and galMOR-treated mice. Spinal cord sections from galMOR-treated mice showed a greater increase in density but not area of MOR-positive staining. These results suggest that using injury-specific promoters to drive gene expression in primary afferent neurons can influence the onset and magnitude of antinociception in a rodent model of neuropathic pain and can be used to upregulate MOR expression in populations of neurons that are potentially injury specific. PERSPECTIVE An injury-specific promoter (galMOR) was used to drive MOR expression in a population- and injury-specific manner. GalMOR increased antinociception and density of MOR staining in the spinal cord. This article presents evidence that promoter selection is an important component in successful gene expression in an injury- and population-specific manner.
Collapse
|
17
|
Nasirinezhad F, Gajavelli S, Priddy B, Jergova S, Zadina J, Sagen J. Viral vectors encoding endomorphins and serine histogranin attenuate neuropathic pain symptoms after spinal cord injury in rats. Mol Pain 2015; 11:2. [PMID: 25563474 PMCID: PMC4349602 DOI: 10.1186/1744-8069-11-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 12/16/2014] [Indexed: 12/22/2022] Open
Abstract
Background The treatment of spinal cord injury (SCI)-induced neuropathic pain presents a challenging healthcare problem. The lack of available robust pharmacological treatments underscores the need for novel therapeutic methods and approaches. Due to the complex character of neuropathic pain following SCI, therapies targeting multiple mechanisms may be a better choice for obtaining sufficient long-term pain relief. Previous studies in our lab showed analgesic effects using combinations of an NMDA antagonist peptide [Ser1]histogranin (SHG), and the mu-opioid peptides endomorphins (EMs), in several pain models. As an alternative to drug therapy, this study evaluated the analgesic potential of these peptides when delivered via gene therapy. Results Lentiviruses encoding SHG and EM-1 and EM-2 were intraspinally injected, either singly or in combination, into rats with clip compression SCI 2 weeks following injury. Treated animals showed significant reduction in mechanical and thermal hypersensitivity, compared to control groups injected with GFP vector only. The antinociceptive effects of individually injected components were modest, but the combination of EMs and SHG produced robust and sustained antinociception. The onset of the analgesic effects was observed between 1–5 weeks post-injection and sustained without decrement for at least 7 weeks. No adverse effects on locomotor function were observed. The involvement of SHG and EMs in the observed antinociception was confirmed by pharmacologic inhibition using intrathecal injection of either the opioid antagonist naloxone or an anti-SHG antibody. Immunohistochemical analysis showed the presence of SHG and EMs in the spinal cord of treated animals, and immunodot-blot analysis of CSF confirmed the presence of these peptides in injected animals. In a separate group of rats, delayed injection of viral vectors was performed in order to mimic a more likely clinical scenario. Comparable and sustained antinociceptive effects were observed in these animals using the SHG-EMs combination vectors compared to the group with early intervention. Conclusions Findings from this study support the potential for direct gene therapy to provide a robust and sustained alleviation of chronic neuropathic pain following SCI. The combination strategy utilizing potent mu-opioid peptides with a naturally-derived NMDA antagonist may produce additive or synergistic analgesic effects without the tolerance development for long-term management of persistent pain.
Collapse
Affiliation(s)
| | | | | | | | | | - Jacqueline Sagen
- Miami Project to Cure Paralysis, University of Miami Miller School Of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
18
|
Protease-Activated Receptor 2 Antagonist Potentiates Analgesic Effects of Systemic Morphine in a Rat Model of Bone Cancer Pain. Reg Anesth Pain Med 2015; 40:158-65. [DOI: 10.1097/aap.0000000000000211] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Glorioso JC. Herpes simplex viral vectors: late bloomers with big potential. Hum Gene Ther 2014; 25:83-91. [PMID: 24502405 DOI: 10.1089/hum.2014.2501] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Joseph C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine , Pittsburgh, PA 15219
| |
Collapse
|
20
|
Viet CT, Dang D, Ye Y, Ono K, Campbell RR, Schmidt BL. Demethylating drugs as novel analgesics for cancer pain. Clin Cancer Res 2014; 20:4882-4893. [PMID: 24963050 DOI: 10.1158/1078-0432.ccr-14-0901] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE In this study, we evaluated the analgesic potential of demethylating drugs on oral cancer pain. Although demethylating drugs could affect expression of many genes, we focused on the mu-opioid receptor (OPRM1) gene pathway, because of its role in pain processing. We determined the antinociceptive effect of OPRM1 re-expression in a mouse oral cancer model. EXPERIMENTAL DESIGN Using a mouse oral cancer model, we determined whether demethylating drugs produced antinociception through re-expression of OPRM1. We then re-expressed OPRM1 with adenoviral transduction and determined if, and by what mechanism, OPRM1 re-expression produced antinociception. To determine the clinical significance of OPRM1 on cancer pain, we quantified OPRM1 methylation in painful cancer tissues and nonpainful contralateral normal tissues of patients with oral cancer, and nonpainful dysplastic tissues of patients with oral dysplasia. RESULTS We demonstrated that OPRM1 was methylated in cancer tissue, but not normal tissue, of patients with oral cancer, and not in dysplastic tissues from patients with oral dysplasia. Treatment with demethylating drugs resulted in mechanical and thermal antinociception in the mouse cancer model. This behavioral change correlated with OPRM1 re-expression in the cancer and associated neurons. Similarly, adenoviral-mediated OPRM1 re-expression on cancer cells resulted in naloxone-reversible antinociception. OPRM1 re-expression on oral cancer cells in vitro increased β-endorphin secretion from the cancer, and decreased activation of neurons that were treated with cancer supernatant. CONCLUSION Our study establishes the regulatory role of methylation in cancer pain. OPRM1 re-expression in cancer cells produces antinociception through cancer-mediated endogenous opioid secretion. Demethylating drugs have an analgesic effect that involves OPRM1.
Collapse
Affiliation(s)
- Chi T Viet
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| | - Dongmin Dang
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| | - Yi Ye
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| | - Kentaro Ono
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| | - Ronald R Campbell
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| | - Brian L Schmidt
- Department of Oral Maxillofacial Surgery, New York University, New York, NY, United States.,Bluestone Center for Clinical Research, New York University, NY, United States
| |
Collapse
|
21
|
Guedon JMG, Zhang M, Glorioso JC, Goins WF, Kinchington PR. Relief of pain induced by varicella-zoster virus in a rat model of post-herpetic neuralgia using a herpes simplex virus vector expressing enkephalin. Gene Ther 2014; 21:694-702. [PMID: 24830437 DOI: 10.1038/gt.2014.43] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 03/24/2014] [Accepted: 03/27/2014] [Indexed: 11/09/2022]
Abstract
Acute and chronic pain (post-herpetic neuralgia or PHN) are encountered in patients with herpes zoster that is caused by reactivation of varicella-zoster virus (VZV) from a state of neuronal latency. PHN is often refractory to current treatments, and additional strategies for pain relief are needed. Here we exploited a rat footpad model of PHN to show that herpes simplex virus (HSV) vector-mediated gene delivery of human preproenkephalin (vHPPE) effectively reduced chronic VZV-induced nocifensive indicators of pain. VZV inoculated at the footpad induced prolonged mechanical allodynia and thermal hyperalgesia that did not develop in controls or with ultraviolet light-inactivated VZV. Subsequent footpad administration of vHPPE relieved VZV-induced pain behaviors in a dose-dependent manner for extended periods, and prophylactic vector administration prevented VZV-induced pain from developing. Short-term pain relief following low-dose vHPPE administration could be effectively prolonged by vector re-administration. HPPE transcripts were increased three- to fivefold in ipsilateral ganglia, but not in the contralateral dorsal root ganglia. VZV hypersensitivity and its relief by vHPPE were not affected by peripheral delivery of opioid receptor agonist or antagonist, suggesting that the efficacy was mediated at the ganglion and/or spinal cord level. These results support further development of ganglionic expression of enkephalin as a novel treatment for the pain associated with Zoster.
Collapse
Affiliation(s)
- J-M G Guedon
- 1] Graduate Program in Molecular Virology and Microbiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA [2] Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - M Zhang
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - J C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - W F Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - P R Kinchington
- 1] Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA [2] Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
22
|
Huang W, Zheng W, Ouyang H, Yi H, Liu S, Zeng W, Levitt RC, Candiotti KA, Lubarsky DA, Hao S. Mechanical allodynia induced by nucleoside reverse transcriptase inhibitor is suppressed by p55TNFSR mediated by herpes simplex virus vector through the SDF1α/CXCR4 system in rats. Anesth Analg 2014; 118:671-80. [PMID: 24557113 DOI: 10.1213/ane.0000000000000079] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND In the human immunodeficiency virus (HIV)-associated sensory neuropathy, neuropathic pain associated with the use of nucleoside reverse transcriptase inhibitors (NRTIs) in patients with HIV/acquired immunodeficiency syndrome is clinically common. While evidence demonstrates that neuropathic pain is influenced by neuroinflammatory events that include the proinflammatory molecules, tumor necrosis factor-α (TNF-α), stromal cell-derived factor 1-α (SDF1-α), and C-X-C chemokine receptor type 4 (CXCR4), the detailed mechanisms by which NRTIs contribute to the development of neuropathic pain are not known. In this study, we investigated the role of these proinflammatory molecules in the dorsal root ganglion (DRG) and the spinal dorsal horn in NRTIs-mediated neuropathic pain state. METHODS Neuropathic pain was induced by intraperitoneal administration of 2',3'-dideoxycytidine (ddC, one of the NRTIs). Mechanical threshold was tested using von Frey filament fibers. Nonreplicating herpes simplex virus (HSV) vectors expressing p55 TNF soluble receptor (p55TNFSR) were inoculated into hindpaw of rats. The expression of TNF-α, SDF1-α, and CXCR4 in both the lumbar spinal cord and the L4/5 DRG was examined using Western blots. Intrathecal CXCR4 antagonist was administered. RESULTS The present study demonstrated that (1) systemic ddC induced upregulation of TNF-α, SDF1-α, and CXCR4 in both the lumbar spinal cord and the L4/5 DRG; (2) p55TNFSR mediated by a nonreplicating HSV vector reversed mechanical allodynia induced by systemic ddC; (3) intrathecal administration of the CXCR4 antagonist AMD3100 increased mechanical threshold; and (4) HSV vector expressing p55TNFSR reversed upregulation of TNF-α, SDF1-α, and CXCR4 induced by ddC in the lumbar spinal dorsal horn and the DRG. CONCLUSIONS Our studies demonstrate that TNF-α through the SDF1/CXCR4 system is involved in the NRTIs-related neuropathic pain state and that blocking the signaling of these proinflammatory molecules is able to reduce NRTIs-related neuropathic pain. These results provide a novel mechanism-based approach (gene therapy) to treating HIV-associated neuropathic pain.
Collapse
Affiliation(s)
- Wan Huang
- From the *Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida; †Department of Anesthesiology, State Key Laboratory of Oncology on Southern China, Cancer Center, Sun Yat-Sen University, Guangzhou, China; ‡Department of Neurology, University of Michigan Medical School, Ann Arbor, Michigan; §Hussman Institute of Human Genomics, University of Miami Miller School of Medicine; and ‖Veterans Affairs Medical Center, Miami, Florida
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Huang W, Zheng W, Liu S, Zeng W, Levitt RC, Candiotti KA, Lubarsky DA, Hao S. HSV-mediated p55TNFSR reduces neuropathic pain induced by HIV gp120 in rats through CXCR4 activity. Gene Ther 2014; 21:328-36. [DOI: 10.1038/gt.2013.90] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 11/26/2013] [Accepted: 11/28/2013] [Indexed: 12/19/2022]
|
24
|
Goss JR, Krisky D, Wechuck J, Wolfe D. Herpes simplex virus-based nerve targeting gene therapy in pain management. J Pain Res 2014; 7:71-9. [PMID: 24470772 PMCID: PMC3901742 DOI: 10.2147/jpr.s36619] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chronic pain represents a major medical burden not only in terms of suffering but also in terms of economic costs. Traditional medical approaches have so far proven insufficient in treating chronic pain and new approaches are necessary. Gene therapy with herpes simplex virus (HSV)-based vectors offers the ability to directly target specific regions of the neuraxis involved in pain transmission including the primary afferent nociceptor. This opens up new targets to interact with that are either not available to traditional systemic drugs or cannot be adequately acted upon without substantial adverse off-target effects. Having access to the entire neuron, which HSV-based vector gene therapy enables, expands treatment options beyond merely treating symptoms and allows for altering the basic biology of the nerve. In this paper, we discuss several HSV-based gene therapy vectors that our group and others have used to target specific neuronal functions involved in the processing of nociception in order to develop new therapies for the treatment of chronic pain.
Collapse
|
25
|
Kantor B, Bailey RM, Wimberly K, Kalburgi SN, Gray SJ. Methods for gene transfer to the central nervous system. ADVANCES IN GENETICS 2014; 87:125-97. [PMID: 25311922 DOI: 10.1016/b978-0-12-800149-3.00003-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gene transfer is an increasingly utilized approach for research and clinical applications involving the central nervous system (CNS). Vectors for gene transfer can be as simple as an unmodified plasmid, but more commonly involve complex modifications to viruses to make them suitable gene delivery vehicles. This chapter will explain how tools for CNS gene transfer have been derived from naturally occurring viruses. The current capabilities of plasmid, retroviral, adeno-associated virus, adenovirus, and herpes simplex virus vectors for CNS gene delivery will be described. These include both focal and global CNS gene transfer strategies, with short- or long-term gene expression. As is described in this chapter, an important aspect of any vector is the cis-acting regulatory elements incorporated into the vector genome that control when, where, and how the transgene is expressed.
Collapse
Affiliation(s)
- Boris Kantor
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina, Columbia, SC, USA
| | - Rachel M Bailey
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Keon Wimberly
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sahana N Kalburgi
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Steven J Gray
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Ophthalmology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
26
|
Reichling DB, Green PG, Levine JD. The fundamental unit of pain is the cell. Pain 2013; 154 Suppl 1:S2-9. [PMID: 23711480 DOI: 10.1016/j.pain.2013.05.037] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 04/12/2013] [Accepted: 05/20/2013] [Indexed: 12/22/2022]
Abstract
The molecular/genetic era has seen the discovery of a staggering number of molecules implicated in pain mechanisms [18,35,61,69,96,133,150,202,224]. This has stimulated pharmaceutical and biotechnology companies to invest billions of dollars to develop drugs that enhance or inhibit the function of many these molecules. Unfortunately this effort has provided a remarkably small return on this investment. Inevitably, transformative progress in this field will require a better understanding of the functional links among the ever-growing ranks of "pain molecules," as well as their links with an even larger number of molecules with which they interact. Importantly, all of these molecules exist side-by-side, within a functional unit, the cell, and its adjacent matrix of extracellular molecules. To paraphrase a recent editorial in Science magazine [223], although we live in the Golden age of Genetics, the fundamental unit of biology is still arguably the cell, and the cell is the critical structural and functional setting in which the function of pain-related molecules must be understood. This review summarizes our current understanding of the nociceptor as a cell-biological unit that responds to a variety of extracellular inputs with a complex and highly organized interaction of signaling molecules. We also discuss the insights that this approach is providing into peripheral mechanisms of chronic pain and sex dependence in pain.
Collapse
Affiliation(s)
- David B Reichling
- Department of Medicine, Division of Neuroscience, University of California-San Francisco, San Francisco, CA, USA; Department of Oral and Maxillofacial Surgery, Division of Neuroscience, University of California-San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
27
|
Molet J, Pohl M. Gene-based approaches in pain research and exploration of new therapeutic targets and strategies. Eur J Pharmacol 2013; 716:129-41. [PMID: 23500201 DOI: 10.1016/j.ejphar.2013.01.073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 01/17/2013] [Accepted: 01/29/2013] [Indexed: 12/18/2022]
Abstract
Large panel of gene-based techniques is used for many years specifically in the pain research field. From the first identification (cloning) of some "mythic" genes, such as those encoding opioid or capsaicin receptors allowing then the creation of first-generation knockout mice, to the today conditional (time, tissue, cell-type and even pathology-dependent) and regulatable modulation of a gene function, these approaches largely contributed to fundamental leaps forward in our understanding of the function of some proteins and of their interest as possible druggable targets. Perhaps one of the most remarkable evolution in the last years is the passage of these approaches from the bench to the patient; whether it concerns the identification of genes involved in inherited pain insensibility/susceptibility, the search for genetic markers of pain types, the individual pharmacogenomics or even the first gene therapy trials. From many possible variants of gene-grounded techniques used in pain research we focus here on gene knockouts and some recent developments, on viral vectors-based gene transfer and on transgenic models for the tracing of pain pathways. Through these selected examples we attempted to emphasize the immense potential of these approaches and their already well-recognized contribution in both the basic and clinical pain research.
Collapse
Affiliation(s)
- Jenny Molet
- INSERM UMRS 975, CNRS UMR 7225, UPMC, Equipe Douleurs , Faculté de Médecine Pitié-Salpêtrière, 91 Bd de l'Hôpital, 75013 Paris, France.
| | | |
Collapse
|
28
|
Wu F, Xu X, Miao X, Chen J, Sun Y, Yu W. Effect of recombinant adenovirus coding for endomorphin-2 on neuropathic pain in rats. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2012; 5:914-923. [PMID: 23119108 PMCID: PMC3484491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Accepted: 09/24/2012] [Indexed: 06/01/2023]
Abstract
OBJECTIVE To construct a transgene expressing human endomorphin-2 by linking the signal peptide of mouse nerve growth factor (PN) to a human endomorphin-2 DNA sequence containing a short linker recognized by the protease FURIN and test the analgesic effect of endomorphin-2 on neuropathic pain. METHODS The transgene was inserted into the cosmid pAxCAwt to generate PN-EM-2-pAxCAwt. The recombinant adenovirus Ad-PNEM2 was packaged and propagated in HEK293 cells. After the Ad-PNEM2-infected NIH3T3 cells had been cultured, protein expression was examined by immunofluorescence and ELISA. A CCI rat model was constructed and the Ad-PNEM2 was administered intrathecally. The rats' pain thresholds (PWL) were measured and the presence of endomorphin-2 in the cerebrospinal fluid was confirmed through ELISA. RESULTS The Ad-PNEM2 expressed endomorphin-2 smoothly and abundantly in NIH3T3 cells at a significantly higher rate than the viral control (P<0.01) or blank control (P<0.01). The expressed endomorphin-2 was mainly observed in the cytoplasm. The concentration of endomorphin-2 in the cerebrospinal fluid increased 1 day after injection and peaked between 7 and 14 days after injection. After injection, PWL approached normal levels in the operated study group. No significant change was observed in the control groups. There was a significant correlation between PWL and endomorphin-2 level (r = 0.944, P<0.001). CONCLUSION The constructed human endomorphin-2 transgene was expressed effectively, and endomorphin-2 expressed by the recombinant adenovirus altered the threshold to thermal stimulus and showed good analgesic effect.
Collapse
Affiliation(s)
- Feixiang Wu
- Department of Anesthesiology, Eastern Hepatobiliary HospitalShanghai 200433, China
| | - Xuewu Xu
- Department of Anesthesiology, Hospital 306Beijing 100101, China
| | - Xuerong Miao
- Department of Anesthesiology, Eastern Hepatobiliary HospitalShanghai 200433, China
| | - Jiaying Chen
- Department of Anesthesiology, Eastern Hepatobiliary HospitalShanghai 200433, China
| | - Yuming Sun
- Department of Anesthesiology, Eastern Hepatobiliary HospitalShanghai 200433, China
| | - Weifeng Yu
- Department of Anesthesiology, Eastern Hepatobiliary HospitalShanghai 200433, China
| |
Collapse
|
29
|
Goins WF, Cohen JB, Glorioso JC. Gene therapy for the treatment of chronic peripheral nervous system pain. Neurobiol Dis 2012; 48:255-70. [PMID: 22668775 DOI: 10.1016/j.nbd.2012.05.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 05/11/2012] [Accepted: 05/24/2012] [Indexed: 11/30/2022] Open
Abstract
Chronic pain is a major health concern affecting 80 million Americans at some time in their lives with significant associated morbidity and effects on individual quality of life. Chronic pain can result from a variety of inflammatory and nerve damaging events that include cancer, infectious diseases, autoimmune-related syndromes and surgery. Current pharmacotherapies have not provided an effective long-term solution as they are limited by drug tolerance and potential abuse. These concerns have led to the development and testing of gene therapy approaches to treat chronic pain. The potential efficacy of gene therapy for pain has been reported in numerous pre-clinical studies that demonstrate pain control at the level of the spinal cord. This promise has been recently supported by a Phase-I human trial in which a replication-defective herpes simplex virus (HSV) vector was used to deliver the human pre-proenkephalin (hPPE) gene, encoding the natural opioid peptides met- and leu-enkephalin (ENK), to cancer patients with intractable pain resulting from bone metastases (Fink et al., 2011). The study showed that the therapy was well tolerated and that patients receiving the higher doses of therapeutic vector experienced a substantial reduction in their overall pain scores for up to a month post vector injection. These exciting early clinical results await further patient testing to demonstrate treatment efficacy and will likely pave the way for other gene therapies to treat chronic pain.
Collapse
Affiliation(s)
- William F Goins
- Dept of Microbiology & Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA 15219, USA.
| | | | | |
Collapse
|
30
|
Wolfe D, Mata M, Fink DJ. Targeted drug delivery to the peripheral nervous system using gene therapy. Neurosci Lett 2012; 527:85-9. [PMID: 22565023 DOI: 10.1016/j.neulet.2012.04.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 04/20/2012] [Indexed: 10/28/2022]
Abstract
Gene transfer to target delivery of neurotrophic factors to the primary sensory afferent for treatment of polyneuropathy, or of inhibitory neurotransmitters for relief of chronic pain, offers the possibility of a highly selective targeted release of bioactive molecules within the nervous system. Preclinical studies with non-replicating herpes simplex virus (HSV)-based vectors injected into the skin to transduce neurons in the dorsal root ganglion have demonstrated efficacy in reducing-pain related behaviors in animal models of inflammatory pain, neuropathic pain, and pain caused by cancer, and in preventing progression of sensory neuropathy caused by toxins, chemotherapeutic drugs or resulting from diabetes. Successful completion of the first phase 1 clinical trial of HSV-mediated gene transfer in patients with intractable pain from cancer has set the stage for further clinical trials of this approach.
Collapse
|
31
|
Zou W, Huang C, Yang Y, Pan Y, Yan J, Guo Q. Microinjection of HSV-1 Amplicon Vector-Mediated Human Proenkephalin into the Periaqueductal Grey Attenuates Neuropathic Pain in Rats. Int J Neurosci 2011; 122:189-94. [DOI: 10.3109/00207454.2011.637653] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
Zheng X, Ouyang H, Liu S, Mata M, Fink DJ, Hao S. TNFα is involved in neuropathic pain induced by nucleoside reverse transcriptase inhibitor in rats. Brain Behav Immun 2011; 25:1668-76. [PMID: 21741472 PMCID: PMC3191308 DOI: 10.1016/j.bbi.2011.06.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 06/13/2011] [Accepted: 06/14/2011] [Indexed: 12/11/2022] Open
Abstract
In patients with HIV/AIDS, neuropathic pain is a common neurological complication. Infection with the HIV itself may lead to neuropathic pain, and painful symptoms are enhanced when patients are treated with nucleoside reverse transcriptase inhibitors (NRTIs). The mechanisms by which NRTIs contribute to the development of neuropathic pain are not known. In the current studies, we tested the role of TNFα in antiretroviral drug-induced neuropathic pain. We administered 2',3'-dideoxycytidine (ddC, one of the NRTIs) systemically to induce mechanical allodynia. We found that ddC induced overexpression of both mRNA and proteins of GFAP and TNFα in the spinal dorsal horn. TNFα was colocalized with GFAP in the spinal dorsal horn and with NeuN in the DRG. Knockdown of TNFα with siRNA blocked the mechanical allodynia induced by ddC. Intrathecal administration of glial inhibitor or recombinant TNF soluble receptor, reversed mechanical allodynia induced by ddC. These results suggest that TNFα is involved in NRTI-induced neuropathic pain.
Collapse
Affiliation(s)
- Xuexing Zheng
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL33136,College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, Jilin Province, P. R. China
| | - Handong Ouyang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - Shue Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL33136
| | - Marina Mata
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - David J. Fink
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - Shuanglin Hao
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL33136,Correspondence to: Dr. Shuanglin Hao, Associate Professor, Research, Department of Anesthesiology, University of Miami Miller School of Medicine, 1550 NW 10th Avenue, Fox BLDG, Rm304C, Miami, FL 33136, Tel: 1-305-243-6420, Fax: 1-305-243-9160,
| |
Collapse
|
33
|
Chattopadhyay M, Mata M, Fink DJ. Vector-mediated release of GABA attenuates pain-related behaviors and reduces Na(V)1.7 in DRG neurons. Eur J Pain 2011; 15:913-920. [PMID: 21486703 PMCID: PMC3321314 DOI: 10.1016/j.ejpain.2011.03.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 03/03/2011] [Accepted: 03/17/2011] [Indexed: 01/15/2023]
Abstract
Pain is a common and debilitating accompaniment of neuropathy that occurs as a complication of diabetes. In the current study, we examined the effect of continuous release of gamma amino butyric acid (GABA), achieved by gene transfer of glutamic acid decarboxylase (GAD67) to dorsal root ganglia (DRG) in vivo using a non-replicating herpes simplex virus (HSV)-based vector (vG) in a rat model of painful diabetic neuropathy (PDN). Subcutaneous inoculation of vG reduced mechanical hyperalgesia, thermal hyperalgesia and cold allodynia in rats with PDN. Continuous release of GABA from vector transduced cells in vivo prevented the increase in the voltage-gated sodium channel isoform 1.7 (Na(V)1.7) protein that is characteristic of PDN. In vitro, infection of primary DRG neurons with vG prevented the increase in Na(V)1.7 resulting from exposure to hyperglycemia. The effect of vector-mediated GABA on Na(V)1.7 levels in vitro was blocked by phaclofen but not by bicuculline, a GABA(B) receptor effect that was blocked by pertussis toxin-(PTX) interference with Gα((i/o)) function. Taken in conjunction with our previous observation that continuous activation of delta opioid receptors by vector-mediated release of enkephalin also prevents the increase in Na(V)1.7 in DRG exposed to hyperglycemia in vitro or in vivo, the observations in this report suggest a novel common mechanism through which activation of G protein coupled receptors (GPCR) in DRG neurons regulate the phenotype of the primary afferent.
Collapse
Affiliation(s)
- Munmun Chattopadhyay
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | | | | |
Collapse
|
34
|
Affiliation(s)
- David J Fink
- University of Michigan and Ann Arbor VA Healthcare System, Ann Arbor MI and Diamyd Inc Pittsburgh PA
| | | |
Collapse
|
35
|
Fink DJ, Wechuck J, Mata M, Glorioso JC, Goss J, Krisky D, Wolfe D. Gene therapy for pain: results of a phase I clinical trial. Ann Neurol 2011; 70:207-12. [PMID: 21796661 DOI: 10.1002/ana.22446] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 04/01/2011] [Accepted: 04/01/2011] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Preclinical evidence indicates that gene transfer to the dorsal root ganglion using replication-defective herpes simplex virus (HSV)-based vectors can reduce pain-related behavior in animal models of pain. This clinical trial was carried out to assess the safety and explore the potential efficacy of this approach in humans. METHODS We conducted a multicenter, dose-escalation, phase I clinical trial of NP2, a replication-defective HSV-based vector expressing human preproenkephalin (PENK) in subjects with intractable focal pain caused by cancer. NP2 was injected intradermally into the dermatome(s) corresponding to the radicular distribution of pain. The primary outcome was safety. As secondary measures, efficacy of pain relief was assessed using a numeric rating scale (NRS), the Short Form McGill Pain Questionnaire (SF-MPQ), and concurrent opiate usage. RESULTS Ten subjects with moderate to severe intractable pain despite treatment with >200mg/day of morphine (or equivalent) were enrolled into the study. Treatment was well tolerated with no study agent-related serious adverse events observed at any point in the study. Subjects receiving the low dose of NP2 reported no substantive change in pain. Subjects in the middle- and high-dose cohorts reported pain relief as assessed by NRS and SF-MPQ. INTERPRETATION Treatment of intractable pain with NP2 was well tolerated. There were no placebo controls in this relatively small study, but the dose-responsive analgesic effects suggest that NP2 may be effective in reducing pain and warrants further clinical investigation.
Collapse
Affiliation(s)
- David J Fink
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, Ann Arbor, MI, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
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
|
37
|
Huang Y, Liu X, Dong L, Liu Z, He X, Liu W. Development of viral vectors for gene therapy for chronic pain. PAIN RESEARCH AND TREATMENT 2011; 2011:968218. [PMID: 22110937 PMCID: PMC3200086 DOI: 10.1155/2011/968218] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 01/31/2011] [Indexed: 11/17/2022]
Abstract
Chronic pain is a major health concern that affects millions of people. There are no adequate long-term therapies for chronic pain sufferers, leading to significant cost for both society and the individual. The most commonly used therapy for chronic pain is the application of opioid analgesics and nonsteroidal anti-inflammatory drugs, but these drugs can lead to addiction and may cause side effects. Further studies of the mechanisms of chronic pain have opened the way for development of new treatment strategies, one of which is gene therapy. The key to gene therapy is selecting safe and highly efficient gene delivery systems that can deliver therapeutic genes to overexpress or suppress relevant targets in specific cell types. Here we review several promising viral vectors that could be applied in gene transfer for the treatment of chronic pain and further discuss the possible mechanisms of genes of interest that could be delivered with viral vectors for the treatment of chronic pain.
Collapse
Affiliation(s)
- Yu Huang
- School of Medicine, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| | - Xin Liu
- College of Pharmacy, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| | - Lanlan Dong
- School of Medicine, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| | - Zhongchun Liu
- School of Medicine, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| | - Xiaohua He
- School of Medicine, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
- Research Center of Food and Drug Evaluation, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| | - Wanhong Liu
- School of Medicine, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
- Research Center of Food and Drug Evaluation, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| |
Collapse
|
38
|
Abstract
This paper is the 32nd consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2009 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, NY 11367, USA.
| |
Collapse
|
39
|
Hui R, Wang W, Chen T, Lü BC, Li H, Zhang T, Wu SX, Li YQ. Origins of endomorphin-2 immunopositive fibers and terminals in the spinal dorsal horn of the rat. Neuroscience 2010; 169:422-30. [PMID: 20457220 DOI: 10.1016/j.neuroscience.2010.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 05/02/2010] [Accepted: 05/03/2010] [Indexed: 01/06/2023]
Abstract
Endomorphin 2 (EM2) plays essential roles in regulating nociceptive transmission within the spinal dorsal horn, where EM2-immunopositive (EM2-IP) fibers and terminals are densely encountered. However, the origins of these EM2-IP structures are still obscure. Unilateral primary sensory afferents disruption (lumbar 3-6 dorsal roots rhizotomy) significantly decreased the density of EM2-IP fibers and terminals in the superficial laminae (laminae I and II) on the ipsilateral but not contralateral lumbar dorsal horn (LDH). Spinal hemisection at the 7th thoracic (T7) segment down-regulated bilateral EM2 expression, with a higher influence on the ipsilateral side of the LDH. Unilateral L3-6 dorsal roots rhizotomy combined with spinal transection but not with hemisection at T7 level completely obliterated EM2-IP fibers and terminals on the rhizotomized-side of the LDH. Disruption of bilateral (exposure to the primary afferent neurotoxin, capsaicin) primary sensory afferents combined with spinal hemisection at T7 decreased the EM2-IP density bilaterally but could obliterate it on neither side of the LDH. While in capsaicin plus transection rats, EM2 was depleted symmetrically and completely. In the colchicine treated rats, no EM2-IP neuronal cell bodies could be detected in the spinal gray matter. After injecting tetramethyl rhodamine dextran-amine (TMR) into the LDH, some of the TMR retrogradely labeled neurons in the nucleus tractus solitarii (NTS) showed EM2-immunoreactivities. The present results indicate that EM2-IP fibers and terminals in the spinal dorsal horn originate from the ipsilateral primary afferents and bilateral descending fibers from NTS.
Collapse
Affiliation(s)
- R Hui
- Department of Anatomy, Histology & Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, PR China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Peripheral non-viral MIDGE vector-driven delivery of beta-endorphin in inflammatory pain. Mol Pain 2009; 5:72. [PMID: 20003437 PMCID: PMC2797781 DOI: 10.1186/1744-8069-5-72] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Accepted: 12/14/2009] [Indexed: 01/13/2023] Open
Abstract
Background Leukocytes infiltrating inflamed tissue produce and release opioid peptides such as β-endorphin, which activate opioid receptors on peripheral terminals of sensory nerves resulting in analgesia. Gene therapy is an attractive strategy to enhance continuous production of endogenous opioids. However, classical viral and plasmid vectors for gene delivery are hampered by immunogenicity, recombination, oncogene activation, anti-bacterial antibody production or changes in physiological gene expression. Non-viral, non-plasmid minimalistic, immunologically defined gene expression (MIDGE) vectors may overcome these problems as they carry only elements needed for gene transfer. Here, we investigated the effects of a nuclear localization sequence (NLS)-coupled MIDGE encoding the β-endorphin precursor proopiomelanocortin (POMC) on complete Freund's adjuvant-induced inflammatory pain in rats. Results POMC-MIDGE-NLS injected into inflamed paws appeared to be taken up by leukocytes resulting in higher concentrations of β-endorphin in these cells. POMC-MIDGE-NLS treatment reversed enhanced mechanical sensitivity compared with control MIDGE-NLS. However, both effects were moderate, not always statistically significant or directly correlated with each other. Also, the anti-hyperalgesic actions could not be increased by enhancing β-endorphin secretion or by modifying POMC-MIDGE-NLS to code for multiple copies of β-endorphin. Conclusion Although MIDGE vectors circumvent side-effects associated with classical viral and plasmid vectors, the current POMC-MIDGE-NLS did not result in reliable analgesic effectiveness in our pain model. This was possibly associated with insufficient and variable efficacy in transfection and/or β-endorphin production. Our data point at the importance of the reproducibility of gene therapy strategies for the control of chronic pain.
Collapse
|
41
|
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
|
42
|
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
|