1
|
Sharma A, Behl T, Sharma L, Shah OP, Yadav S, Sachdeva M, Rashid S, Bungau SG, Bustea C. Exploring the molecular pathways and therapeutic implications of angiogenesis in neuropathic pain. Biomed Pharmacother 2023; 162:114693. [PMID: 37062217 DOI: 10.1016/j.biopha.2023.114693] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/26/2023] [Accepted: 04/10/2023] [Indexed: 04/18/2023] Open
Abstract
Recently, much attention has been paid to chronic neuro-inflammatory condition underlying neuropathic pain. It is generally linked with thermal hyperalgesia and tactile allodynia. It results due to injury or infection in the nervous system. The neuropathic pain spectrum covers a variety of pathophysiological states, mostly involved are ischemic injury viral infections associated neuropathies, chemotherapy-induced peripheral neuropathies, autoimmune disorders, traumatic origin, hereditary neuropathies, inflammatory disorders, and channelopathies. In CNS, angiogenesis is evident in inflammation of neurons and pain in bone cancer. The role of chemokines and cytokines is dualistic; their aggressive secretion produces detrimental effects, leading to neuropathic pain. However, whether the angiogenesis contributes and exists in neuropathic pain remains doubtful. In the present review, we elucidated summary of diverse mechanisms of neuropathic pain associated with angiogenesis. Moreover, an overview of multiple targets that have provided insights on the VEGF signaling, signaling through Tie-1 and Tie-2 receptor, erythropoietin pathway promoting axonal growth are also discussed. Because angiogenesis as a result of these signaling, results in inflammation, we focused on the mechanisms of neuropathic pain. These factors are mainly responsible for the activation of post-traumatic regeneration of the PNS and CNS. Furthermore, we also reviewed synthetic and herbal treatments targeting angiogenesis in neuropathic pain.
Collapse
Affiliation(s)
- Aditi Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan 173211, Himachal Pradesh, India
| | - Tapan Behl
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, 248007 Dehradun, Uttarakhand, India.
| | - Lalit Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan 173211, Himachal Pradesh, India
| | - Om Prakash Shah
- School of Pharmaceutical Sciences, Shoolini University, Solan 173211, Himachal Pradesh, India
| | - Shivam Yadav
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Chhatrapati Shahu ji Maharaj University, Kanpur 208024, Uttar Pradesh, India
| | - Monika Sachdeva
- Fatima College of Health Sciences, Al Ain 00000, United Arab Emirates
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea 410028, Romania; Doctoral School of Biomedical Sciences, University of Oradea, Oradea 410028, Romania.
| | - Cristiana Bustea
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, Oradea 410073, Romania
| |
Collapse
|
2
|
Jia S, Wei G, Bono J, Pan Z, Zheng B, Wang B, Adaralegbe A, Tenorio C, Bekker A, Tao YX. TET1 overexpression attenuates paclitaxel-induced neuropathic pain through rescuing K 2p1.1 expression in primary sensory neurons of male rats. Life Sci 2022; 297:120486. [PMID: 35304127 PMCID: PMC8976761 DOI: 10.1016/j.lfs.2022.120486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/03/2022] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
AIMS Paclitaxel-induced downregulation of two-pore domain K+ channel 1.1 (K2p1.1) caused by increasing DNA methylation within its gene promoter in the dorsal root ganglion (DRG) contributes to neuropathic pain. Given that ten-eleven translocation methylcytosine dioxygenase 1 (TET1) promotes DNA demethylation and gene transcription, the present study investigated whether DRG overexpression of TET1 produces an antinociceptive effect on the paclitaxel-induced nociceptive hypersensitivity. MAIN METHODS TET1 was overexpressed in the DRG through unilateral microinjection of the herpes simplex virus expressing full-length Tet1 mRNA into the fourth and fifth lumbar DRGs of male rats. Behavioral tests were carried out to examine the effect of this overexpression on the paclitaxel-induced nociceptive hypersensitivity. Western blot analysis, chromatin immunoprecipitation assay and 5-hydroxymethylcytosine detection assay were performed to assess the levels of TET1/K2p1.1, 5-methylcytosine and 5-hydroxymethylcytosine, respectively. KEY FINDINGS DRG overexpression of TET1 mitigated the paclitaxel-induced mechanical allodynia, heat hyperalgesia and cold hyperalgesia on the ipsilateral side during the development and maintenance periods. Locomotor function or basal (acute) responses to mechanical, heat or cold stimuli were not affected. Mechanistically, DRG overexpression of TET1 rescued the expression of K2p1.1 by blocking the paclitaxel-induced increase in the level of 5-methylcytosine and correspondingly reversing the paclitaxel-induced decreases in the amount of 5-hydroxymethylcytosine within the K2p1.1 promoter region in the microinjected DRGs of male rats. SIGNIFICANCE Our findings suggest that DRG overexpression of TET1 alleviated chemotherapy-induced neuropathic pain likely through rescuing DRG K2p1.1 expression. Our findings may provide a potential avenue for the management of this disorder.
Collapse
Affiliation(s)
- Shushan Jia
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Guihua Wei
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Jamie Bono
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA,Rutgers School of Graduate Studies, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ07103, USA
| | - Zhiqiang Pan
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Bixin Zheng
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Bing Wang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Adejuyigbe Adaralegbe
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Christopher Tenorio
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Alex Bekker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Cell Biology & Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark NJ07103, USA; Department of Physiology, Pharmacology & Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark NJ07103, USA.
| |
Collapse
|
3
|
Emerging roles of lncRNAs in the pathogenesis, diagnosis, and treatment of trigeminal neuralgia. Biochem Soc Trans 2022; 50:1013-1023. [PMID: 35437600 DOI: 10.1042/bst20220070] [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: 01/27/2022] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 02/05/2023]
Abstract
Trigeminal neuralgia (TN) is one of the most common neuropathic pain disorders and is often combined with other comorbidities if managed inadequately. However, the present understanding of its pathogenesis at the molecular level remains lacking. Long noncoding RNAs (lncRNAs) play crucial roles in neuropathic pain, and many studies have reported that specific lncRNAs are related to TN. This review summarizes the current understanding of lncRNAs in the pathogenesis, diagnosis, and treatment of TN. Recent studies have shown that the lncRNAs uc.48+, Gm14461, MRAK009713 and NONRATT021972 are potential candidate loci for the diagnosis and treatment of TN. The current diagnostic system could be enhanced and improved by a workflow for selecting transcriptomic biomarkers and the development of lncRNA-based molecular diagnostic systems for TN. The discovery of lncRNAs potentially impacts drug selection for TN; however, the current supporting evidence is limited to preclinical studies. Additional studies are needed to further test the diagnostic and therapeutic value of lncRNAs in TN.
Collapse
|
4
|
Askarian-Amiri S, Maleki SN, Alavi SNR, Neishaboori AM, Toloui A, Gubari MIM, Sarveazad A, Hosseini M, Yousefifard M. The efficacy of GABAergic precursor cells transplantation in alleviating neuropathic pain in animal models: a systematic review and meta-analysis. Korean J Pain 2022; 35:43-58. [PMID: 34966011 PMCID: PMC8728544 DOI: 10.3344/kjp.2022.35.1.43] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 12/11/2022] Open
Abstract
Background Current therapies are quite unsuccessful in the management of neuropathic pain. Therefore, considering the inhibitory characteristics of GABA mediators, the present systematic review and meta-analysis aimed to determine the efficacy of GABAergic neural precursor cells on neuropathic pain management. Methods Search was conducted on Medline, Embase, Scopus, and Web of Science databases. A search strategy was designed based on the keywords related to GABAergic cells combined with neuropathic pain. The outcomes were allodynia and hyperalgesia. The results were reported as a pooled standardized mean difference (SMD) with a 95% confidence interval (95% CI). Results Data of 13 studies were analyzed in the present meta-analysis. The results showed that administration of GABAergic cells improved allodynia (SMD = 1.79; 95% CI 0.87, 271; P < 0.001) and hyperalgesia (SMD = 1.29; 95% CI 0.26, 2.32; P = 0.019). Moreover, the analyses demonstrated that the efficacy of GABAergic cells in the management of allodynia and hyperalgesia is only observed in rats. Also, only genetically modified cells are effective in improving both of allodynia, and hyperalgesia. Conclusions A moderate level of pre-clinical evidence showed that transplantation of genetically-modified GABAergic cells is effective in the management of neuropathic pain. However, it seems that the transplantation efficacy of these cells is only statistically significant in improving pain symptoms in rats. Hence, caution should be exercised regarding the generalizability and the translation of the findings from rats and mice studies to large animal studies and clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Amirmohammad Toloui
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammed I M Gubari
- Department of Family and Community Medicine, College of Medicine, University of Sulaimani, Sulaimani, Iraq
| | - Arash Sarveazad
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran.,Nursing Care Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mostafa Hosseini
- Pediatric Chronic Kidney Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Yousefifard
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Khare P, Dave KM, Kamte YS, Manoharan MA, O'Donnell LA, Manickam DS. Development of Lipidoid Nanoparticles for siRNA Delivery to Neural Cells. AAPS J 2021; 24:8. [PMID: 34873640 PMCID: PMC8648339 DOI: 10.1208/s12248-021-00653-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/28/2021] [Indexed: 01/24/2023] Open
Abstract
Lipidoid nanoparticles (LNPs) are the delivery platform in Onpattro, the first FDA-approved siRNA drug. LNPs are also the carriers in the Pfizer-BioNTech and Moderna COVID-19 mRNA vaccines. While these applications have demonstrated that LNPs effectively deliver nucleic acids to hepatic and muscle cells, it is unclear if LNPs could be used for delivery of siRNA to neural cells, which are notoriously challenging delivery targets. Therefore, the purpose of this study was to determine if LNPs could efficiently deliver siRNA to neurons. Because of their potential delivery utility in either applications for the central nervous system and the peripheral nervous system, we used both cortical neurons and sensory neurons. We prepared siRNA-LNPs using C12-200, a benchmark ionizable cationic lipidoid along with helper lipids. We demonstrated using dynamic light scattering that the inclusion of both siRNA and PEG-lipid provided a stabilizing effect to the LNP particle diameters and polydispersity indices by minimizing aggregation. We found that siRNA-LNPs were safely tolerated by primary dorsal root ganglion neurons. Flow cytometry analysis revealed that Cy5 siRNA delivered via LNPs into rat primary cortical neurons showed uptake levels similar to Lipofectamine RNAiMAX-the gold standard commercial transfection agent. However, LNPs demonstrated a superior safety profile, whereas the Lipofectamine-mediated uptake was concomitant with significant toxicity. Fluorescence microscopy demonstrated a time-dependent increase in the uptake of LNP-delivered Cy5 siRNA in a human cortical neuron cell line. Overall, our results suggest that LNPs are a viable platform that can be optimized for delivery of therapeutic siRNAs to neural cells.
Collapse
Affiliation(s)
- Purva Khare
- Graduate School of Pharmaceutical Sciences, Duquesne University, 453 Mellon Hall, 600 Forbes Avenue, Pittsburgh, Pennsylvania, 15282, USA
| | - Kandarp M Dave
- Graduate School of Pharmaceutical Sciences, Duquesne University, 453 Mellon Hall, 600 Forbes Avenue, Pittsburgh, Pennsylvania, 15282, USA
| | - Yashika S Kamte
- Graduate School of Pharmaceutical Sciences, Duquesne University, 453 Mellon Hall, 600 Forbes Avenue, Pittsburgh, Pennsylvania, 15282, USA
| | | | - Lauren A O'Donnell
- Graduate School of Pharmaceutical Sciences, Duquesne University, 453 Mellon Hall, 600 Forbes Avenue, Pittsburgh, Pennsylvania, 15282, USA
| | - Devika S Manickam
- Graduate School of Pharmaceutical Sciences, Duquesne University, 453 Mellon Hall, 600 Forbes Avenue, Pittsburgh, Pennsylvania, 15282, USA.
| |
Collapse
|
6
|
Katiyar N, Raju G, Madhusudanan P, Gopalakrishnan-Prema V, Shankarappa SA. Neuronal delivery of nanoparticles via nerve fibres in the skin. Sci Rep 2021; 11:2566. [PMID: 33510229 PMCID: PMC7844288 DOI: 10.1038/s41598-021-81995-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/13/2021] [Indexed: 11/13/2022] Open
Abstract
Accessing the peripheral nervous system (PNS) by topically applied nanoparticles is a simple and novel approach with clinical applications in several PNS disorders. Skin is richly innervated by long peripheral axons that arise from cell bodies located distally within ganglia. In this study we attempt to target dorsal root ganglia (DRG) neurons, via their axons by topical application of lectin-functionalized gold nanoparticles (IB4-AuNP). In vitro, 140.2 ± 1.9 nm IB4-AuNP were found to bind both axons and cell bodies of DRG neurons, and AuNP applied at the axonal terminals were found to translocate to the cell bodies. Topical application of IB4-AuNP on rat hind-paw resulted in accumulation of three to fourfold higher AuNP in lumbar DRG than in contralateral control DRGs. Results from this study clearly suggest that topically applied nanoparticles with neurotropic targeting ligands can be utilized for delivering nanoparticles to neuronal cell bodies via axonal transport mechanisms.
Collapse
Affiliation(s)
- Neeraj Katiyar
- Center for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham University, Kochi, 682041, Kerala, India
| | - Gayathri Raju
- Center for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham University, Kochi, 682041, Kerala, India
| | - Pallavi Madhusudanan
- Center for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham University, Kochi, 682041, Kerala, India
| | - Vignesh Gopalakrishnan-Prema
- Center for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham University, Kochi, 682041, Kerala, India
| | - Sahadev A Shankarappa
- Center for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham University, Kochi, 682041, Kerala, India.
| |
Collapse
|
7
|
Yang Y, Wen J, Zheng B, Wu S, Mao Q, Liang L, Li Z, Bachmann T, Bekker A, Tao YX. CREB Participates in Paclitaxel-Induced Neuropathic Pain Genesis Through Transcriptional Activation of Dnmt3a in Primary Sensory Neurons. Neurotherapeutics 2021; 18:586-600. [PMID: 33051852 PMCID: PMC8116406 DOI: 10.1007/s13311-020-00931-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2020] [Indexed: 12/16/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathic pain (CIPNP) often occurs in cancer patients treated with antineoplastic drugs. Therapeutic management of CIPNP is very limited, at least in part due to the largely unknown mechanisms that underlie CIPNP genesis. Here, we showed that systemic administration of the chemotherapeutic drug paclitaxel significantly and time-dependently increased the levels of cyclic AMP response element-binding protein (CREB) in dorsal root ganglion (DRG) neurons. Blocking this increase through DRG microinjection of Creb siRNA attenuated paclitaxel-induced mechanical, heat, and cold nociceptive hypersensitivities. Mimicking this increase through DRG microinjection of the adeno-associated virus 5 expressing full-length Creb mRNA led to enhanced responses to basal mechanical, heat, and cold stimuli in mice in absence of paclitaxel treatment. Mechanically, paclitaxel-induced increase of DRG CREB protein augmented Dnmt3a promoter activity and participated in the paclitaxel-induced upregulation of DNMT3a protein in the DRG. CREB overexpression also elevated the expression of DNMT3a in in vivo and in vitro DRG neurons of naïve mice. Given that DNMT3a is an endogenous instigator of CIPNP and that CREB co-expresses with DNMT3a in DRG neurons, CREB may be a key player in CIPNP through transcriptional activation of the Dnmt3a gene in primary sensory neurons. CREB is thus a likely potential target for the therapeutic management of this disorder.
Collapse
Affiliation(s)
- Yong Yang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Jing Wen
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Bixin Zheng
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Shaogen Wu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Qingxiang Mao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Lingli Liang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Zhisong Li
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Thomas Bachmann
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Alex Bekker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA.
- Department of Physiology, Pharmacology & Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA.
- Department of Cell Biology & Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA.
| |
Collapse
|
8
|
Mody PH, Pathak S, Hanson LK, Spencer JV. Herpes Simplex Virus: A Versatile Tool for Insights Into Evolution, Gene Delivery, and Tumor Immunotherapy. Virology (Auckl) 2020; 11:1178122X20913274. [PMID: 34093008 PMCID: PMC8142529 DOI: 10.1177/1178122x20913274] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 02/19/2020] [Indexed: 12/20/2022] Open
Abstract
Herpesviruses are prevalent throughout the animal kingdom, and they have coexisted and coevolved along with their host species for millions of years. Herpesviruses carry a large (120-230 kb) double-stranded DNA genome surrounded by a protein capsid, a tegument layer consisting of viral and host proteins, and a lipid bilayer envelope with surface glycoproteins. A key characteristic of these viruses is their ability to enter a latent state following primary infection, allowing them to evade the host's immune system and persist permanently. Herpesviruses can reactivate from their dormant state, usually during times of stress or when the host's immune responses are impaired. While herpesviruses can cause complications with severe disease in immune-compromised people, most of the population experiences few ill effects from herpesvirus infections. Indeed, herpes simplex virus 1 (HSV-1) in particular has several features that make it an attractive tool for therapeutic gene delivery. Herpes simplex virus 1 targets and infects specific cell types, such as epithelial cells and neurons. The HSV-1 genome can also accommodate large insertions of up to 14 kb. The HSV-1-based vectors have already achieved success for the oncolytic treatment of melanoma. In addition to serving as a vehicle for therapeutic gene delivery and targeted cell lysis, comparative genomics of herpesviruses HSV-1 and 2 has revealed valuable information about the evolutionary history of both viruses and their hosts. This review focuses on the adaptability of HSV-1 as an instrument for gene delivery and an evolutionary marker. Overall, HSV-1 shows great promise as a tool for treating human disease and studying human migration patterns, disease outbreaks, and evolution.
Collapse
Affiliation(s)
- Prapti H Mody
- Department of Biology, Texas Woman’s University, Denton, TX, USA
| | - Sushila Pathak
- Department of Biology, Texas Woman’s University, Denton, TX, USA
| | - Laura K Hanson
- Department of Biology, Texas Woman’s University, Denton, TX, USA
| | - Juliet V Spencer
- Department of Biology, Texas Woman’s University, Denton, TX, USA
| |
Collapse
|
9
|
Wu Q, Wei G, Ji F, Jia S, Wu S, Guo X, He L, Pan Z, Miao X, Mao Q, Yang Y, Cao M, Tao YX. TET1 Overexpression Mitigates Neuropathic Pain Through Rescuing the Expression of μ-Opioid Receptor and Kv1.2 in the Primary Sensory Neurons. Neurotherapeutics 2019; 16:491-504. [PMID: 30515739 PMCID: PMC6554405 DOI: 10.1007/s13311-018-00689-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Peripheral nerve injury downregulates the expression of the μ-opioid receptor (MOR) and voltage-gated potassium channel subunit Kv1.2 by increasing their DNA methylation in the dorsal root ganglion (DRG). Ten-eleven translocation methylcytosine dioxygenase 1 (TET1) causes DNA demethylation. Given that DRG MOR and Kv1.2 downregulation contribute to neuropathic pain genesis, this study investigated the effect of DRG TET1 overexpression on neuropathic pain. Overexpression of TET1 in the DRG through microinjection of herpes simplex virus expressing full-length TET1 mRNA into the injured rat DRG significantly alleviated the fifth lumbar spinal nerve ligation (SNL)-induced pain hypersensitivities during the development and maintenance periods, without altering acute pain or locomotor function. This microinjection also restored morphine analgesia and attenuated morphine analgesic tolerance development after SNL. Mechanistically, TET1 microinjection rescued the expression of MOR and Kv1.2 by reducing the level of 5-methylcytosine and increasing the level of 5-hydroxymethylcytosine in the promoter and 5' untranslated regions of the Oprml1 gene (encoding MOR) and in the promoter region of the Kcna2 gene (encoding Kv1.2) in the DRG ipsilateral to SNL. These findings suggest that DRG TET1 overexpression mitigated neuropathic pain likely through rescue of MOR and Kv1.2 expression in the ipsilateral DRG. Virus-mediated DRG delivery of TET1 may open a new avenue for neuropathic pain management.
Collapse
Affiliation(s)
- Qiang Wu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Guihua Wei
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
- Department of Pharmacy, Institute of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Fengtao Ji
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Shushan Jia
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
- Department of Anesthesiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Shaogen Wu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
| | - Xinying Guo
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
| | - Long He
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
| | - Zhiqiang Pan
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
| | - Xuerong Miao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
| | - Qingxiang Mao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
| | - Yong Yang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA
| | - Minghui Cao
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China.
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 S. Orange Ave., MSB, E-661, Newark, NJ, 07103, USA.
| |
Collapse
|
10
|
Wu S, Bono J, Tao YX. Long noncoding RNA (lncRNA): a target in neuropathic pain. Expert Opin Ther Targets 2018; 23:15-20. [PMID: 30451044 DOI: 10.1080/14728222.2019.1550075] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Current treatments for neuropathic pain are limited in part due to the incomplete understanding of its underlying mechanisms. Recent evidence reveals the dysregulated expression of long non-coding RNAs (lncRNAs) in the damaged nerve, dorsal root ganglion (DRG), and spinal cord dorsal horn following peripheral nerve injury. However, the role of the majority of lncRNAs in neuropathic pain genesis is still elusive. Unveiling the mechanisms of how lncRNAs participate in neuropathic pain may develop new strategies to prevent and/or treat this disorder. Areas covered: This review focuses on the dysregulation of lncRNAs in the DRG, dorsal horn, and the injured nerves from preclinical models of neuropathic pain. We provide evidence of how peripheral nerve injury causes the dysregulation of lncRNAs in these pain-related regions. The potential mechanisms of how dysregulated lncRNAs contribute to the pathogenesis of neuropathic pain are discussed. Expert opinion: The investigation on the role of the dysregulated lncRNAs in neuropathic pain might open up a novel avenue for therapeutic treatment of this disorder. However, current investigation is at the infancy stage, which challenges the translation of preclinical findings. More intensive studies on lncRNAs are required before the preclinical findings are translated into therapeutic management for neuropathic pain.
Collapse
Affiliation(s)
- Shaogen Wu
- a Department of Anesthesiology , New Jersey Medical School, Rutgers, The State University of New Jersey , Newark , NJ , USA
| | - Jamie Bono
- a Department of Anesthesiology , New Jersey Medical School, Rutgers, The State University of New Jersey , Newark , NJ , USA
| | - Yuan-Xiang Tao
- a Department of Anesthesiology , New Jersey Medical School, Rutgers, The State University of New Jersey , Newark , NJ , USA
| |
Collapse
|
11
|
Langert KA, Brey EM. Strategies for Targeted Delivery to the Peripheral Nerve. Front Neurosci 2018; 12:887. [PMID: 30542262 PMCID: PMC6277764 DOI: 10.3389/fnins.2018.00887] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/13/2018] [Indexed: 12/17/2022] Open
Abstract
Delivery of compounds to the peripheral nervous system has the potential to be used as a treatment for a broad range of conditions and applications, including neuropathic pain, regional anesthesia, traumatic nerve injury, and inherited and inflammatory neuropathies. However, efficient delivery of therapeutic doses can be difficult to achieve due to peripheral neuroanatomy and the restrictiveness of the blood-nerve barrier. Depending on the underlying integrity of the blood-nerve barrier in the application at hand, several strategies can be employed to navigate the peripheral nerve architecture and facilitate targeted delivery to the peripheral nerve. This review describes different applications where targeted delivery to the peripheral nervous system is desired, the challenges that the blood-nerve barrier poses in each application, and bioengineering strategies that can facilitate delivery in each application.
Collapse
Affiliation(s)
- Kelly A Langert
- Department of Veterans Affairs, Research Service, Edward Hines, Jr. VA Hospital, Hines, IL, United States.,Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, United States
| | - Eric M Brey
- Audie L. Murphy VA Hospital, San Antonio, TX, United States.,Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
12
|
Willemen HLDM, Kavelaars A, Prado J, Maas M, Versteeg S, Nellissen LJJ, Tromp J, Gonzalez Cano R, Zhou W, Jakobsson ME, Małecki J, Posthuma G, Habib AM, Heijnen CJ, Falnes PØ, Eijkelkamp N. Identification of FAM173B as a protein methyltransferase promoting chronic pain. PLoS Biol 2018; 16:e2003452. [PMID: 29444090 PMCID: PMC5828452 DOI: 10.1371/journal.pbio.2003452] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 02/27/2018] [Accepted: 01/24/2018] [Indexed: 12/31/2022] Open
Abstract
Chronic pain is a debilitating problem, and insights in the neurobiology of chronic pain are needed for the development of novel pain therapies. A genome-wide association study implicated the 5p15.2 region in chronic widespread pain. This region includes the coding region for FAM173B, a functionally uncharacterized protein. We demonstrate here that FAM173B is a mitochondrial lysine methyltransferase that promotes chronic pain. Knockdown and sensory neuron overexpression strategies showed that FAM173B is involved in persistent inflammatory and neuropathic pain via a pathway dependent on its methyltransferase activity. FAM173B methyltransferase activity in sensory neurons hyperpolarized mitochondria and promoted macrophage/microglia activation through a reactive oxygen species–dependent pathway. In summary, we uncover a role for methyltransferase activity of FAM173B in the neurobiology of pain. These results also highlight FAM173B methyltransferase activity as a potential therapeutic target to treat debilitating chronic pain conditions. Pain is an evolutionarily conserved physiological phenomenon necessary for survival. Yet, pain can become pathological when it occurs independently of noxious stimuli. The molecular mechanisms of pathological pain are still poorly understood, limiting the development of highly needed novel analgesics. Recently, genetic variations in the genomic region encoding FAM173B—a functionally uncharacterized protein—have been linked to chronic pain in humans. In this study, we identify the role and function of FAM173B in the development of pathological pain. We used genetic, biochemical, and behavioral approaches in mice to show that FAM173B is a mitochondrial lysine methyltransferase—a protein that transfers methyl group to donor proteins. By genetically silencing or overexpressing FAM173B in sensory neurons, we showed that FAM173B methyltransferase activity promotes the development of chronic pain. In addition, we discovered that FAM173B methyltransferase activity in the mitochondria of sensory neurons promotes chronic pain via a pathway that depends on the production of reactive oxygen species and on the engagement of spinal cord microglia—engulfing cells of the central nervous system. These data point to an essential role of FAM173B in the regulation of pathological pain.
Collapse
Affiliation(s)
- Hanneke L. D. M. Willemen
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Annemieke Kavelaars
- Laboratory of Neuroimmunology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Judith Prado
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Mirjam Maas
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sabine Versteeg
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lara J. J. Nellissen
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jeshua Tromp
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Rafael Gonzalez Cano
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Pharmacology and Institute of Neuroscience, University of Granada, Granada, Spain
| | - Wenjun Zhou
- Laboratory of Neuroimmunology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Magnus E. Jakobsson
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Jędrzej Małecki
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - George Posthuma
- Department of Cell Biology and Institute of Biomembranes, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Abdella M. Habib
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
- College of Medicine, Member of Qatar Health, Qatar University, Doha, Qatar
| | - Cobi J. Heijnen
- Laboratory of Neuroimmunology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Pål Ø. Falnes
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Niels Eijkelkamp
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- * E-mail:
| |
Collapse
|
13
|
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
|
14
|
Meidahl AC, Klukinov M, Tzabazis AZ, Sorensen JC, Yeomans DC. Nasal application of HSV encoding human preproenkephalin blocks craniofacial pain in a rat model of traumatic brain injury. Gene Ther 2017; 24:482-486. [PMID: 28682314 DOI: 10.1038/gt.2017.55] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/06/2017] [Accepted: 06/14/2017] [Indexed: 12/18/2022]
Abstract
According to Centers for Disease Control and Prevention, each year, an estimated 1.7 million Americans sustain a traumatic brain injury (TBI), which frequently leads to chronic craniofacial pain. In this study we examine a gene therapy approach to the treatment of post-TBI craniofacial neuropathic pain using nasal application of a herpes simplex virus (HSV)-based vector expressing human proenkephalin (SHPE) to target the trigeminal ganglia. Mild TBI was induced in rats by the use of a modified fluid percussion model. Two days after mild TBI, following the development of facial mechanical allodynia, animals received either an intranasal application of vehicle or recombinant HSV encoding human preproenkephalin or lacZ reporter gene encoding control vector (SHZ.1). Compared with baseline response thresholds, mild TBI in SHZ.1 or vehicle-treated animals induced a robust craniofacial allodynia lasting at least 45 days. On the other hand, nasal SHPE application 2 days post-TBI attenuated facial allodynia, reaching significance by day 4-7 and maintaining this effect throughout the duration of the experiment. Immunohistochemical examination revealed strong expression of human proenkephalin in trigeminal ganglia of SHPE, but not SHZ.1-treated rats. This study demonstrates that intranasal administration of HSV-based gene vectors may be a viable, non-invasive means of treating chronic craniofacial pain, including post-TBI pain.
Collapse
Affiliation(s)
- A C Meidahl
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
| | - M Klukinov
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - A Z Tzabazis
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - J C Sorensen
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
| | - D C Yeomans
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
15
|
Spahn V, Stein C. Targeting delta opioid receptors for pain treatment: drugs in phase I and II clinical development. Expert Opin Investig Drugs 2017; 26:155-160. [PMID: 28001096 DOI: 10.1080/13543784.2017.1275562] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Opioids are widely used to treat severe pain. Most clinically used opioids activate µ-opioid receptors (MOR). Their ligands induce potent analgesia but also adverse effects. The δ-opioid receptor (DOR) is another member of the opioid receptor family that has been under intense investigation with the aim to avoid MOR-induced side effects. Areas covered: This article reviews DOR ligands which appeared to be promising after preclinical evaluation. A literature search using Pubmed, Cochrane library, ClinicalTrials.gov, EudraCT, AdisInsight database and EBSCO Online Library was conducted. Out of numerous newly synthesized molecules, only few candidates entered phase I and/or II clinical investigation. The publicly accessible results are presented here. Expert opinion: Many compounds showed potent DOR-specific pain inhibition in preclinical studies. ADL5859 and ADL5747 entered clinical trials and successfully passed phase I. However, in phase II studies the primary endpoint (pain reduction) was not met and further investigation was terminated. A third compound, NP2, is in phase II clinical evaluation and results are pending. These findings suggest a potential of DOR ligands according to preclinical studies. Further clinical research and secondary analysis of unpublished data is needed to identify molecules which are useful in humans.
Collapse
Affiliation(s)
- Viola Spahn
- a Klinik für Anästhesiologie und operative Intensivmedizin , Charité Campus Benjamin Franklin, Freie Universität Berlin , Berlin , Germany
| | - Christoph Stein
- a Klinik für Anästhesiologie und operative Intensivmedizin , Charité Campus Benjamin Franklin, Freie Universität Berlin , Berlin , Germany.,b Multifunctional Biomaterials for Medicine , Helmholtz Virtual Institute , Teltow , Germany
| |
Collapse
|
16
|
Johnson T, Koria P. Expression and Purification of Neurotrophin-Elastin-Like Peptide Fusion Proteins for Neural Regeneration. BioDrugs 2016; 30:117-27. [PMID: 26820997 DOI: 10.1007/s40259-016-0159-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Neural injuries such as spinal cord injuries, traumatic brain injuries, or nerve transection injuries pose a major health problem. Neurotrophins such as nerve growth factor (NGF) or brain-derived neurotrophic factor (BDNF) have been shown to improve the outcome of neural injuries in several pre-clinical models, but their use in clinics is limited by the lack of a robust delivery system that enhances their bioavailability and half-life. OBJECTIVES We describe two fusion proteins comprising NGF or BDNF fused with elastin-like peptides (ELPs). The aim of this study was to investigate the biological activity of neurotrophin-ELP (N-ELP) fusion proteins via in vitro culture models. METHODS NGF and BDNF were cloned in front of an elastin-like polypeptide sequence V40C2. These proteins were expressed in bacteria as inclusion bodies. These fusion proteins underwent solubilization via 8 M urea and purification via inverse transition cycling (ITC). We measured the particle size and the effect of temperature on precipitated particles using dynamic light scattering (DLS). We used western blot analysis to confirm the specificity of NGF-ELP to tropomyosin receptor kinase A (TrkA) antibody and to confirm the specificity of BDNF-ELP to TrkB antibody. PC12 cells were used to perform a neurite outgrowth assay to determine the biological activity of NGF-ELP. Bioactivity of BDNF-ELP was ascertained via transfecting human epithelial kidney (HEK 293-T) cells to express the TrkB receptor. RESULTS The proteins were successfully purified to high homogeneity by exploiting the phase transition property of ELPs and urea, which solubilize inclusion bodies. Using PC12 neurite outgrowth assay, we further demonstrated that the biological activity of NGF was retained in the fusion. Similarly, BDNF-ELP phosphorylated the TrkB receptor, suggesting the biological activity of BDNF was also retained in the fusion. We further show that owing to the phase transition property of ELPs in the fusion, these proteins self-assembled into nanoparticles at their respective transition temperatures. CONCLUSION These fusion proteins are useful for neural regeneration, as they not only retain the biological activity of the neurotrophin but also self-assemble into nanoparticles, thereby simultaneously serving as drug-delivery vehicles. These nanoparticles can serve as drug depots and will increase bioavailability by limiting neurotrophin loss due to diffusion, thereby allowing controlled spatio-temporal delivery of the neurotrophin.
Collapse
Affiliation(s)
- Tamina Johnson
- Department of Chemical and Biomedical Engineering, University of South Florida, 4202 E Fowler Avenue, ENB 118, Tampa, FL, 33620, USA
| | - Piyush Koria
- Department of Chemical and Biomedical Engineering, University of South Florida, 4202 E Fowler Avenue, ENB 118, Tampa, FL, 33620, USA.
| |
Collapse
|
17
|
Ischemic optic neuropathy as a model of neurodegenerative disorder: A review of pathogenic mechanism of axonal degeneration and the role of neuroprotection. J Neurol Sci 2016; 375:430-441. [PMID: 28320183 DOI: 10.1016/j.jns.2016.12.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 02/07/2023]
Abstract
Optic neuropathy is a neurodegenerative disease which involves optic nerve injury. It is caused by acute or intermittent insults leading to visual dysfunction. There are number of factors, responsible for optic neuropathy, and the optic nerve axon is affected in all type which causes the loss of retinal ganglion cells. In this review we will highlight various mechanisms involved in the cell loss cascades during axonal degeneration as well as ischemic optic neuropathy. These mechanisms include oxidative stress, excitotoxicity, angiogenesis, neuroinflammation and apoptosis following retinal ischemia. We will also discuss the effect of neuroprotective agents in attenuation of the negative effect of factors involve in the disease occurrence and progression.
Collapse
|
18
|
Chang MF, Hsieh JH, Chiang H, Kan HW, Huang CM, Chellis L, Lin BS, Miaw SC, Pan CL, Chao CC, Hsieh ST. Effective gene expression in the rat dorsal root ganglia with a non-viral vector delivered via spinal nerve injection. Sci Rep 2016; 6:35612. [PMID: 27748450 PMCID: PMC5066268 DOI: 10.1038/srep35612] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 10/04/2016] [Indexed: 12/13/2022] Open
Abstract
Delivering gene constructs into the dorsal root ganglia (DRG) is a powerful but challenging therapeutic strategy for sensory disorders affecting the DRG and their peripheral processes. The current delivery methods of direct intra-DRG injection and intrathecal injection have several disadvantages, including potential injury to DRG neurons and low transfection efficiency, respectively. This study aimed to develop a spinal nerve injection strategy to deliver polyethylenimine mixed with plasmid (PEI/DNA polyplexes) containing green fluorescent protein (GFP). Using this spinal nerve injection approach, PEI/DNA polyplexes were delivered to DRG neurons without nerve injury. Within one week of the delivery, GFP expression was detected in 82.8% ± 1.70% of DRG neurons, comparable to the levels obtained by intra-DRG injection (81.3% ± 5.1%, p = 0.82) but much higher than those obtained by intrathecal injection. The degree of GFP expression by neurofilament(+) and peripherin(+) DRG neurons was similar. The safety of this approach was documented by the absence of injury marker expression, including activation transcription factor 3 and ionized calcium binding adaptor molecule 1 for neurons and glia, respectively, as well as the absence of behavioral changes. These results demonstrated the efficacy and safety of delivering PEI/DNA polyplexes to DRG neurons via spinal nerve injection.
Collapse
Affiliation(s)
- Ming-Fong Chang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Jung-Hsien Hsieh
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Departments of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Hao Chiang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Hung-Wei Kan
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Cho-Min Huang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Luke Chellis
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, USA
| | - Bo-Shiou Lin
- Department of Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Shi-Chuen Miaw
- Department of Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chun-Liang Pan
- Department of Graduate Institute of Molecular Medicine, College of Medicine, National Taiwan University, No. 7 Chung-Shan South Road, Taipei, 10002, Taiwan
| | - Chi-Chao Chao
- Departments of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Graduate Institute of Brain and Mind Science, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Departments of Clinical Center for Neuroscience and Behavior, National Taiwan University Hospital, Taipei, Taiwan
- Department of Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| |
Collapse
|
19
|
Xu Y, Liu J, He M, Liu R, Belegu V, Dai P, Liu W, Wang W, Xia QJ, Shang FF, Luo CZ, Zhou X, Liu S, McDonald J, Liu J, Zuo YX, Liu F, Wang TH. Mechanisms of PDGF siRNA-mediated inhibition of bone cancer pain in the spinal cord. Sci Rep 2016; 6:27512. [PMID: 27282805 PMCID: PMC4901320 DOI: 10.1038/srep27512] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 05/10/2016] [Indexed: 02/05/2023] Open
Abstract
Patients with tumors that metastasize to bone frequently suffer from debilitating pain, and effective therapies for treating bone cancer are lacking. This study employed a novel strategy in which herpes simplex virus (HSV) carrying a small interfering RNA (siRNA) targeting platelet-derived growth factor (PDGF) was used to alleviate bone cancer pain. HSV carrying PDGF siRNA was established and intrathecally injected into the cavum subarachnoidale of animals suffering from bone cancer pain and animals in the negative group. Sensory function was assessed by measuring thermal and mechanical hyperalgesia. The mechanism by which PDGF regulates pain was also investigated by comparing the differential expression of pPDGFRα/β and phosphorylated ERK and AKT. Thermal and mechanical hyperalgesia developed in the rats with bone cancer pain, and these effects were accompanied by bone destruction in the tibia. Intrathecal injection of PDGF siRNA and morphine reversed thermal and mechanical hyperalgesia in rats with bone cancer pain. In addition, we observed attenuated astrocyte hypertrophy, down-regulated pPDGFRα/β levels, reduced levels of the neurochemical SP, a reduction in CGRP fibers and changes in pERK/ERK and pAKT/AKT ratios. These results demonstrate that PDGF siRNA can effectively treat pain induced by bone cancer by blocking the AKT-ERK signaling pathway.
Collapse
Affiliation(s)
- Yang Xu
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jia Liu
- Institute of Neuroscience, Kunming Medical University, Kunming 650031, PR China
| | - Mu He
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ran Liu
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Visar Belegu
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA, International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at Kennedy Krieger Inc., Baltimore, MD, USA
| | - Ping Dai
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Wei Liu
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Wei Wang
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qing-Jie Xia
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Fei-Fei Shang
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Chao-Zhi Luo
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xue Zhou
- Department of Histology, Embryology and Neurobiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Su Liu
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA, International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at Kennedy Krieger Inc., Baltimore, MD, USA
| | - JohnW. McDonald
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA, International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at Kennedy Krieger Inc., Baltimore, MD, USA
| | - Jin Liu
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yun-Xia Zuo
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Fei Liu
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ting-Hua Wang
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
- Institute of Neuroscience, Kunming Medical University, Kunming 650031, PR China
| |
Collapse
|
20
|
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
|
21
|
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]
|
22
|
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
|
23
|
Kramer PR, Umorin M, Bellinger LL. Attenuation of myogenic orofacial nociception and mechanical hypersensitivity by viral mediated enkephalin overproduction in male and female rats. BMC Neurol 2015; 15:34. [PMID: 25885338 PMCID: PMC4369359 DOI: 10.1186/s12883-015-0285-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/25/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Clinical studies have tested the use of an engineered herpes virus to treat pain. We hypothesized that subcutaneous injections of an engineered herpes virus that expresses enkephalin would attenuate orofacial nociception and hypersensitivity in male and female rats by a central mechanism. METHODS Herpes virus was injected subcutaneously around the mouth of male and female rats seventy-two hours before ligatures were placed on the masseter tendon, control treatment groups received either no virus or no ligature. Enkephalin expression was measured and von Frey filament testing and meal duration were utilized to measure mechanical hypersensitivity and the nociceptive response, respectively. Naloxone or naloxone methiodide was administered to rats injected with the enkephalin expressing virus to test if enkephalin was acting peripherally or centrally. RESULTS Ligature significantly lengthened meal duration and reduced the threshold to von Frey filaments for 18 days. Infection with the enkephalin transgene significantly decreased this response for at least 11 days but only in male rats. Virus injection significantly increased expression of enkephalin in the mental nerve that innervates the mouth region, the trigeminal ganglia and the trigeminal nucleus caudalis but no increase was observed in the masseter nerve after virus injection. Naloxone but not naloxone methiodide reversed the response to the enkephaline expressing virus. CONCLUSIONS The data suggests that sex should be a considered when using this virus and that viral transfection of the mental nerve with an enkephalin transgene can reduce nociception and hypersensitivity through a central mechanism.
Collapse
Affiliation(s)
- Phillip R Kramer
- Department of Biomedical Sciences, Texas A&M University Baylor College of Dentistry, 3302 Gaston Avenue, Dallas, TX, 75246, USA.
| | - Mikhail Umorin
- Department of Biomedical Sciences, Texas A&M University Baylor College of Dentistry, 3302 Gaston Avenue, Dallas, TX, 75246, USA.
| | - Larry L Bellinger
- Department of Biomedical Sciences, Texas A&M University Baylor College of Dentistry, 3302 Gaston Avenue, Dallas, TX, 75246, USA.
| |
Collapse
|
24
|
Pleticha J, Jeng-Singh C, Rezek R, Zaibak M, Beutler AS. Intraneural convection enhanced delivery of AAVrh20 for targeting primary sensory neurons. Mol Cell Neurosci 2014; 60:72-80. [PMID: 24769104 DOI: 10.1016/j.mcn.2014.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 02/23/2014] [Accepted: 04/14/2014] [Indexed: 11/19/2022] Open
Abstract
Gene therapy using adeno-associated virus (AAV) is an attractive strategy to treat disorders of the peripheral nervous system (PNS), such as chronic pain or peripheral neuropathies. Although intrathecal (IT) administration of AAV has been the standard in the field for targeting the PNS, it lacks anatomical specificity and results in wide rostro-caudal distribution of the vector. An alternative approach is to deliver AAV directly to the peripheral nerve axon. The present study employed convection-enhanced delivery (CED) of a novel AAV serotype, AAVrh20, expressing enhanced green fluorescent protein (EGFP) into rat sciatic nerve investigating its efficacy, anatomical selectivity, and safety, compared to the IT route. Intraneural CED resulted in transduction confined to the ipsilateral L4 and L5 DRG while IT administration led to promiscuous DRG transduction encompassing the entire lumbar region bilaterally. The transduction rate for intraneural AAV administration was similar to IT delivery (24% for L4 and 31.5% for L5 DRG versus 50% for L4 and 19.5% for L5 DRG). The use of hyperosmotic diluent did not further improve the transduction efficiency. AAVrh20 was superior to reference serotypes previously described to be most active for each route. Intraneural CED of AAV was associated with transient allodynia that resolved spontaneously. These findings establish intraneural CED as an alternative to IT administration for AAV mediated gene transfer to the PNS and, based on a reference rodent model, suggest AAVrh20 as a superior serotype for targeting the PNS.
Collapse
Affiliation(s)
- Josef Pleticha
- Departments of Anesthesiology and Oncology, Mayo Clinic, Rochester, MN, USA
| | | | - Rahaf Rezek
- Departments of Anesthesiology and Oncology, Mayo Clinic, Rochester, MN, USA
| | - Manal Zaibak
- Departments of Anesthesiology and Oncology, Mayo Clinic, Rochester, MN, USA
| | - Andreas S Beutler
- Departments of Anesthesiology and Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
25
|
Abstract
Cancer pain is a serious health problem, and imposes a great burden on the lives of patients and their families. Pain can be associated with delay in treatment, denial of treatment, or failure of treatment. If the pain is not treated properly it may impair the quality of life. Neuropathic cancer pain (NCP) is one of the most complex phenomena among cancer pain syndromes. NCP may result from direct damage to nerves due to acute diagnostic/therapeutic interventions. Chronic NCP is the result of treatment complications or malignancy itself. Although the reason for pain is different in NCP and noncancer neuropathic pain, the pathophysiologic mechanisms are similar. Data regarding neuropathic pain are primarily obtained from neuropathic pain studies. Evidence pertaining to NCP is limited. NCP due to chemotherapeutic toxicity is a major problem for physicians. In the past two decades, there have been efforts to standardize NCP treatment in order to provide better medical service. Opioids are the mainstay of cancer pain treatment; however, a new group of therapeutics called coanalgesic drugs has been introduced to pain treatment. These coanalgesics include gabapentinoids (gabapentin, pregabalin), antidepressants (tricyclic antidepressants, duloxetine, and venlafaxine), corticosteroids, bisphosphonates, N-methyl-D-aspartate antagonists, and cannabinoids. Pain can be encountered throughout every step of cancer treatment, and thus all practicing oncologists must be capable of assessing pain, know the possible underlying pathophysiology, and manage it appropriately. The purpose of this review is to discuss neuropathic pain and NCP in detail, the relevance of this topic, clinical features, possible pathology, and treatments of NCP.
Collapse
Affiliation(s)
- Ece Esin
- Medical Oncology Department, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Suayib Yalcin
- Medical Oncology Department, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
26
|
Augmenting glial cell-line derived neurotrophic factor signaling to treat painful neuropathies. Proc Natl Acad Sci U S A 2014; 111:2060-1. [PMID: 24474803 DOI: 10.1073/pnas.1324047111] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
27
|
Wang H, Heijnen CJ, van Velthoven CTJ, Willemen HLDM, Ishikawa Y, Zhang X, Sood AK, Vroon A, Eijkelkamp N, Kavelaars A. Balancing GRK2 and EPAC1 levels prevents and relieves chronic pain. J Clin Invest 2013; 123:5023-34. [PMID: 24231349 DOI: 10.1172/jci66241] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 09/12/2013] [Indexed: 01/12/2023] Open
Abstract
Chronic pain is a major clinical problem, yet the mechanisms underlying the transition from acute to chronic pain remain poorly understood. In mice, reduced expression of GPCR kinase 2 (GRK2) in nociceptors promotes cAMP signaling to the guanine nucleotide exchange factor EPAC1 and prolongs the PGE2-induced increase in pain sensitivity (hyperalgesia). Here we hypothesized that reduction of GRK2 or increased EPAC1 in dorsal root ganglion (DRG) neurons would promote the transition to chronic pain. We used 2 mouse models of hyperalgesic priming in which the transition from acute to chronic PGE2-induced hyperalgesia occurs. Hyperalgesic priming with carrageenan induced a sustained decrease in nociceptor GRK2, whereas priming with the PKCε agonist ΨεRACK increased DRG EPAC1. When either GRK2 was increased in vivo by viral-based gene transfer or EPAC1 was decreased in vivo, as was the case for mice heterozygous for Epac1 or mice treated with Epac1 antisense oligodeoxynucleotides, chronic PGE2-induced hyperalgesia development was prevented in the 2 priming models. Using the CFA model of chronic inflammatory pain, we found that increasing GRK2 or decreasing EPAC1 inhibited chronic hyperalgesia. Our data suggest that therapies targeted at balancing nociceptor GRK2 and EPAC1 levels have promise for the prevention and treatment of chronic pain.
Collapse
|
28
|
Abstract
A number of agents from diverse pharmacological classes are used to treat neuropathic pain associated with diabetic peripheral neuropathy. Only three of these have regulatory approval for this indication in the U.S. In this focused article, I will discuss selected drugs, newly approved or in development, to treat neuropathic pain in patients with diabetic neuropathy. These will include agonists and antagonists of the transient receptor potential channels, a family of receptor proteins that play a role in the transduction of physical stress; sodium channel isoform specific antagonists; a recently approved dual-action opioid receptor agonist-norepinephrine reuptake inhibitor; gene therapy for neuropathic pain; and anti-nerve growth factor molecules. Mechanisms of action, preclinical supporting data, clinical trial evidence, and adverse effects will be reviewed.
Collapse
Affiliation(s)
- Roy Freeman
- Center for Autonomic and Peripheral Nerve Disorders, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| |
Collapse
|
29
|
Yin X, Yu B, Tang Z, He B, Ren J, Xiao X, Tang W. Bifidobacterium infantis-mediated HSV-TK/GCV suicide gene therapy induces both extrinsic and intrinsic apoptosis in a rat model of bladder cancer. Cancer Gene Ther 2012; 20:77-81. [PMID: 23258087 DOI: 10.1038/cgt.2012.86] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bladder cancer is the most common cancer in the urological tract. Here, we investigated the molecular pathways involved in the apoptosis of rodent bladder cancer, which was treated with Bifidobacterium infantis (BI)-mediated herpes simplex virus thymidine kinase (HSV-TK)/ganciclovir (GCV) suicide gene therapy. We engineered the BI-TK system, which consists of BI with the recombinant plasmid PGEX-1λT carrying HSV-TK (that is, TK) gene. Tumor-bearing rats were randomly divided into three groups and tail vein injected with normal saline (group A), BI/PGEX-1λT (group B) or BI-TK (group C), followed by intraperitoneal injection of GCV. The BI-TK/GCV system (group C) was shown to inhibit tumor growth and caused the apoptosis of tumor cells as assessed by in situ terminal transferase dUTP nick-end labeling assay. While varied extents of apoptosis were detected in tumor cells from all groups, tumor cells treated with BI-TK/GCV system (group C) exhibited the highest level of apoptosis (P < 0.05), consistent with our previous studies. Furthermore, we found that the expression levels of Fas, FasL, Cyt-C and caspase-9 in tumor tissues derived from group C were significantly higher than the other two groups (P < 0.001). Therefore, our results have demonstrated that the BI-TK/GCV therapy system exhibits a sustainable antitumor growth activity in the rodent model of bladder cancer. Mechanistically, both extrinsic and intrinsic apoptosis pathways are involved in the BI-TK/GCV antitumor functionality.
Collapse
Affiliation(s)
- X Yin
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|