1
|
Fang H, Li M, Yang J, Ma S, Zhang L, Yang H, Tang Q, Cao J, Yang W. Repressing iron overload ameliorates central post-stroke pain via the Hdac2-Kv1.2 axis in a rat model of hemorrhagic stroke. Neural Regen Res 2024; 19:2708-2722. [PMID: 38595289 PMCID: PMC11168507 DOI: 10.4103/nrr.nrr-d-23-01498] [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: 09/06/2023] [Revised: 11/21/2023] [Accepted: 02/04/2024] [Indexed: 04/11/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202412000-00027/figure1/v/2024-04-08T165401Z/r/image-tiff Thalamic hemorrhage can lead to the development of central post-stroke pain. Changes in histone acetylation levels, which are regulated by histone deacetylases, affect the excitability of neurons surrounding the hemorrhagic area. However, the regulatory mechanism of histone deacetylases in central post-stroke pain remains unclear. Here, we show that iron overload leads to an increase in histone deacetylase 2 expression in damaged ventral posterolateral nucleus neurons. Inhibiting this increase restored histone H3 acetylation in the Kcna2 promoter region of the voltage-dependent potassium (Kv) channel subunit gene in a rat model of central post-stroke pain, thereby increasing Kcna2 expression and relieving central pain. However, in the absence of nerve injury, increasing histone deacetylase 2 expression decreased Kcna2 expression, decreased Kv current, increased the excitability of neurons in the ventral posterolateral nucleus area, and led to neuropathic pain symptoms. Moreover, treatment with the iron chelator deferiprone effectively reduced iron overload in the ventral posterolateral nucleus after intracerebral hemorrhage, reversed histone deacetylase 2 upregulation and Kv1.2 downregulation, and alleviated mechanical hypersensitivity in central post-stroke pain rats. These results suggest that histone deacetylase 2 upregulation and Kv1.2 downregulation, mediated by iron overload, are important factors in central post-stroke pain pathogenesis and could serve as new targets for central post-stroke pain treatment.
Collapse
Affiliation(s)
- He Fang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jingchen Yang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shunping Ma
- Department of Nutrition, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Li Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hongqi Yang
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, Henan Province, China
| | - Qiongyan Tang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing Cao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
- Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, Henan Province, China
| | - Weimin Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
2
|
Meijs S, Hayward AJ, Gomes Nørgaard Dos Santos Nielsen T, Reidies Bjarkam C, Jensen W. Spared ulnar nerve injury results in increased layer III-VI excitability in the pig somatosensory cortex. Lab Anim (NY) 2024; 53:287-293. [PMID: 39349800 PMCID: PMC11442301 DOI: 10.1038/s41684-024-01440-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/21/2024] [Indexed: 10/04/2024]
Abstract
This study describes cortical recordings in a large animal nerve injury model. We investigated differences in primary somatosensory cortex (S1) hyperexcitability when stimulating injured and uninjured nerves and how different cortical layers contribute to S1 hyperexcitability after spared ulnar nerve injury. We used a multielectrode array to record single-neuron activity in the S1 of ten female Danish landrace pigs. Electrical stimulation of the injured and uninjured nerve evoked brain activity up to 3 h after injury. The peak amplitude and latency of early and late peristimulus time histogram responses were extracted for statistical analysis. Histological investigations determined the layer of the cortex in which each electrode contact was placed. Nerve injury increased the early peak amplitude compared with that of the control group. This difference was significant immediately after nerve injury when the uninjured nerve was stimulated, while it was delayed for the injured nerve. The amplitude of the early peak was increased in layers III-VI after nerve injury compared with the control. In layer III, S1 excitability was also increased compared with preinjury for the early peak. Furthermore, the late peak was significantly larger in layer III than in the other layers in the intervention and control group before and after injury. Thus, the most prominent increase in excitability occurred in layer III, which is responsible for the gain modulation of cortical output through layer V. Therefore, layer III neurons seem to have an important role in altered brain excitability after nerve injury.
Collapse
Affiliation(s)
- Suzan Meijs
- Center for Neuroplasticity and Pain, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| | - Andrew J Hayward
- Center for Neuroplasticity and Pain, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | | - Carsten Reidies Bjarkam
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Neurosurgery, Aalborg University Hospital, Aalborg, Denmark
| | - Winnie Jensen
- Center for Neuroplasticity and Pain, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
3
|
Liu H, Lauzadis J, Gunaratna K, Sipple E, Kaczocha M, Puopolo M. Inhibition of T-Type Calcium Channels With TTA-P2 Reduces Chronic Neuropathic Pain Following Spinal Cord Injury in Rats. THE JOURNAL OF PAIN 2023; 24:1681-1695. [PMID: 37169156 DOI: 10.1016/j.jpain.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/03/2023] [Accepted: 05/02/2023] [Indexed: 05/13/2023]
Abstract
Spinal cord injury (SCI)-induced neuropathic pain (SCI-NP) develops in up to 60 to 70% of people affected by traumatic SCI, leading to a major decline in quality of life and increased risk for depression, anxiety, and addiction. Gabapentin and pregabalin, together with antidepressant drugs, are commonly prescribed to treat SCI-NP, but their efficacy is unsatisfactory. The limited efficacy of current pharmacological treatments for SCI-NP likely reflects our limited knowledge of the underlying mechanism(s) responsible for driving the maintenance of SCI-NP. The leading hypothesis in the field supports a major role for spontaneously active injured nociceptors in driving the maintenance of SCI-NP. Recent data from our laboratory provided additional support for this hypothesis and identified the T-type calcium channels as key players in driving the spontaneous activity of SCI-nociceptors, thus providing a rational pharmacological target to treat SCI-NP. To test whether T-type calcium channels contribute to the maintenance of SCI-NP, male and female SCI and sham rats were treated with TTA-P2 (a blocker of T-type calcium channels) to determine its effects on mechanical hypersensitivity (as measured with the von Frey filaments) and spontaneous ongoing pain (as measured with the conditioned place preference paradigm), and compared them to the effects of gabapentin, a blocker of high voltage-activated calcium channels. We found that both TTA-P2 and gabapentin reduced mechanical hypersensitivity in male and females SCI rats, but surprisingly only TTA-P2 reduced spontaneous ongoing pain in male SCI rats. PERSPECTIVES: SCI-induced neuropathic pain, and in particular the spontaneous ongoing pain component, is notoriously very difficult to treat. Our data provide evidence that inhibition of T-type calcium channels reduces spontaneous ongoing pain in SCI rats, supporting a clinically relevant role for T-type channels in the maintenance of SCI-induced neuropathic pain.
Collapse
Affiliation(s)
- Huilin Liu
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Justas Lauzadis
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Kavindu Gunaratna
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Erin Sipple
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York.
| |
Collapse
|
4
|
Rangel-Galván M, Rangel-Galván V, Rangel-Huerta A. T-type calcium channel modulation by hydrogen sulfide in neuropathic pain conditions. Front Pharmacol 2023; 14:1212800. [PMID: 37529702 PMCID: PMC10387653 DOI: 10.3389/fphar.2023.1212800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/05/2023] [Indexed: 08/03/2023] Open
Abstract
Neuropathic pain can appear as a direct or indirect nerve damage lesion or disease that affects the somatosensory nervous system. If the neurons are damaged or indirectly stimulated, immune cells contribute significantly to inflammatory and neuropathic pain. After nerve injury, peripheral macrophages/spinal microglia accumulate around damaged neurons, producing endogenous hydrogen sulfide (H2S) through the cystathionine-γ-lyase (CSE) enzyme. H2S has a pronociceptive modulation on the Cav3.2 subtype, the predominant Cav3 isoform involved in pain processes. The present review provides relevant information about H2S modulation on the Cav3.2 T-type channels in neuropathic pain conditions. We have discussed that the dual effect of H2S on T-type channels is concentration-dependent, that is, an inhibitory effect is seen at low concentrations of 10 µM and an augmentation effect on T-current at 100 µM. The modulation mechanism of the Cav3.2 channel by H2S involves the direct participation of the redox/Zn2+ affinity site located in the His191 in the extracellular loop of domain I of the channel, involving a group of extracellular cysteines, comprising C114, C123, C128, and C1333, that can modify the local redox environment. The indirect interaction pathways involve the regulation of the Cav3.2 channel through cytokines, kinases, and post-translational regulators of channel expression. The findings conclude that the CSE/H2S/Cav3.2 pathway could be a promising therapeutic target for neuropathic pain disorders.
Collapse
Affiliation(s)
- Maricruz Rangel-Galván
- Biothecnology Department, Metropolitan Polytechnic University of Puebla, Puebla, Puebla, Mexico
| | - Violeta Rangel-Galván
- Nursing and Physiotherapy Department, University of Professional Development, Tijuana, Baja California, Mexico
| | - Alejandro Rangel-Huerta
- Faculty of Computer Science, Meritorious Autonomous University of Puebla, Puebla, Puebla, Mexico
| |
Collapse
|
5
|
Chen W, Wang X, Sun Q, Zhang Y, Liu J, Hu T, Wu W, Wei C, Liu M, Ding Y, Liu D, Chong Y, Wang P, Zhu H, Cui W, Zhang J, Li Q, Yang F. The upregulation of NLRP3 inflammasome in dorsal root ganglion by ten-eleven translocation methylcytosine dioxygenase 2 (TET2) contributed to diabetic neuropathic pain in mice. J Neuroinflammation 2022; 19:302. [PMID: 36527131 PMCID: PMC9756585 DOI: 10.1186/s12974-022-02669-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The nucleotide oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) in dorsal root ganglion (DRG) contributes to pain hypersensitivity in multiple neuropathic pain models, but the function of the NLRP3 in diabetic neuropathic pain (DNP) and the regulation mechanism are still largely unknown. Epigenetic regulation plays a vital role in the controlling of gene expression. Ten-eleven translocation methylcytosine dioxygenase 2 (TET2) is a DNA demethylase that contributes to transcriptional activation. TET2 is also involved in high glucose (HG)-induced pathology. METHODS DNP was induced in mice via the intraperitoneal injection of streptozotocin (STZ) for five consecutive days and the mechanical threshold was evaluated in STZ-diabetic mice by using von Frey hairs. The expression level of the NLRP3 pathway and TET2 in DRG were determined through molecular biology experiments. The regulation of the NLRP3 pathway by TET2 was examined in in vitro and in vivo conditions. RESULTS In the present research, we first established the DNP model and found that NLRP3 pathway was activated in DRG. The treatment of NLRP3 inhibitor MCC950 alleviated the mechanical allodynia of DNP mice. Then we revealed that in STZ-diabetic mice DRG, the genomic DNA was demethylated, and the expression of DNA demethylase TET2 was increased evidently. Using RNA-sequencing analysis, we found that the expression of Txnip, a gene that encodes a thioredoxin-interacting protein (TXNIP) which mediates NLRP3 activation, was elevated in the DRG after STZ treatment. In addition, knocking down of TET2 expression in DRG using TET2-siRNA suppressed the mRNA expression of Txnip and subsequently inhibited the expression/activation of NLRP3 inflammasome in vitro and in vivo as well as relieved the pain sensitivity of DNP animals. CONCLUSION The results suggested that the upregulation of the TXNIP/NLRP3 pathway by TET2 in DRG was involved in the pain hypersensitivity of the DNP model.
Collapse
Affiliation(s)
- Wen Chen
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China ,grid.24695.3c0000 0001 1431 9176International Acupuncture and Moxibustion Innovation Institute, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Xiaotong Wang
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Qingyu Sun
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Yurui Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Jing Liu
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Tingting Hu
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Weihua Wu
- grid.24696.3f0000 0004 0369 153XDepartment of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Chao Wei
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Meng Liu
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Yumeng Ding
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Dianxin Liu
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Yingzi Chong
- grid.24696.3f0000 0004 0369 153XDepartment of Anesthesiology Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070 China
| | - Peipei Wang
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Hongwei Zhu
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
| | - Weihua Cui
- grid.24696.3f0000 0004 0369 153XDepartment of Anesthesiology Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070 China
| | - Jiannan Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China
| | - Qian Li
- grid.24696.3f0000 0004 0369 153XDepartment of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China ,grid.24696.3f0000 0004 0369 153XAdvanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069 China ,grid.24696.3f0000 0004 0369 153XKey Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing, 100069 China
| | - Fei Yang
- grid.24696.3f0000 0004 0369 153XDepartment of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069 China ,grid.24696.3f0000 0004 0369 153XAdvanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069 China
| |
Collapse
|
6
|
Tiwari V, Hemalatha S. Betaine Attenuates Chronic Constriction Injury-Induced Neuropathic Pain in Rats by Inhibiting KIF17-Mediated Nociception. ACS Chem Neurosci 2022; 13:3362-3377. [PMID: 36367842 DOI: 10.1021/acschemneuro.2c00380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Kinesin superfamily proteins transport a diverse range of cargo, including excitatory receptors to the dendrite and axon of a neuron via retrograde and anterograde fashions along microtubules, causing central sensitization and neuropathic pain. In this study, we have performed in silico molecular dynamics simulation to delineate the dynamic interaction of betaine with KIF17, a kinesin protein, known to be involved in neuropathic pain. The results from the molecular dynamics study suggest that the betaine-KIF17 complex is stabilized through hydrogen bonding, polar interactions, and water bridges. Findings from in vivo studies suggest a significant increase in pain hypersensitivity, oxido-nitrosative stress, and KIF17 overexpression in the sciatic nerve, dorsal root ganglion (DRG), and spinal cord of nerve-injured rats, which was significantly attenuated on treatment with betaine. Betaine treatment also restored the increased NR2B expressions and levels of proinflammatory cytokines and neuropeptides in the DRG and spinal cord of nerve-injured rats. Findings from the current study suggest that betaine attenuates neuropathic pain in rats by inhibiting KIF17-NR2B-mediated neuroinflammatory signaling.
Collapse
Affiliation(s)
- Vineeta Tiwari
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Siva Hemalatha
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| |
Collapse
|
7
|
Shin SM, Lauzadis J, Itson-Zoske B, Cai Y, Fan F, Natarajan GK, Kwok WM, Puopolo M, Hogan QH, Yu H. Targeting intrinsically disordered regions facilitates discovery of calcium channels 3.2 inhibitory peptides for adeno-associated virus-mediated peripheral analgesia. Pain 2022; 163:2466-2484. [PMID: 35420557 PMCID: PMC9562599 DOI: 10.1097/j.pain.0000000000002650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/19/2022] [Accepted: 03/23/2022] [Indexed: 11/27/2022]
Abstract
ABSTRACT Ample data support a prominent role of peripheral T-type calcium channels 3.2 (Ca V 3.2) in generating pain states. Development of primary sensory neuron-specific inhibitors of Ca V 3.2 channels is an opportunity for achieving effective analgesic therapeutics, but success has been elusive. Small peptides, especially those derived from natural proteins as inhibitory peptide aptamers (iPAs), can produce highly effective and selective blockade of specific nociceptive molecular pathways to reduce pain with minimal off-target effects. In this study, we report the engineering of the potent and selective iPAs of Ca V 3.2 from the intrinsically disordered regions (IDRs) of Ca V 3.2 intracellular segments. Using established prediction algorithms, we localized the IDRs in Ca V 3.2 protein and identified several Ca V 3.2iPA candidates that significantly reduced Ca V 3.2 current in HEK293 cells stably expressing human wide-type Ca V 3.2. Two prototype Ca V 3.2iPAs (iPA1 and iPA2) derived from the IDRs of Ca V 3.2 intracellular loops 2 and 3, respectively, were expressed selectively in the primary sensory neurons of dorsal root ganglia in vivo using recombinant adeno-associated virus (AAV), which produced sustained inhibition of calcium current conducted by Ca V 3.2/T-type channels and significantly attenuated both evoked and spontaneous pain behavior in rats with neuropathic pain after tibial nerve injury. Recordings from dissociated sensory neurons showed that AAV-mediated Ca V 3.2iPA expression suppressed neuronal excitability, suggesting that Ca V 3.2iPA treatment attenuated pain by reversal of injury-induced neuronal hypersensitivity. Collectively, our results indicate that Ca V 3.2iPAs are promising analgesic leads that, combined with AAV-mediated delivery in anatomically targeted sensory ganglia, have the potential to be a selective peripheral Ca V 3.2-targeting strategy for clinical treatment of pain.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Justas Lauzadis
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Yongsong Cai
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Fan Fan
- Department of Pharmacology and Toxicology, The University of Mississippi Medical Center, Jackson, MS, United States
| | - Gayathri K. Natarajan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| | - Quinn H. Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
8
|
Zhi YR, Cao F, Su XJ, Gao SW, Zheng HN, Jiang JY, Su L, Liu J, Wang Y, Zhang Y, Zhang Y. The T-Type Calcium Channel Cav3.2 in Somatostatin Interneurons in Spinal Dorsal Horn Participates in Mechanosensation and Mechanical Allodynia in Mice. Front Cell Neurosci 2022; 16:875726. [PMID: 35465611 PMCID: PMC9024096 DOI: 10.3389/fncel.2022.875726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Somatostatin-positive (SOM+) neurons have been proposed as one of the key populations of excitatory interneurons in the spinal dorsal horn involved in mechanical pain. However, the molecular mechanism for their role in pain modulation remains unknown. Here, we showed that the T-type calcium channel Cav3.2 was highly expressed in spinal SOM+ interneurons. Colocalization of Cacna1h (which codes for Cav3.2) and SOMtdTomato was observed in the in situ hybridization studies. Fluorescence-activated cell sorting of SOMtdTomato cells in spinal dorsal horn also proved a high expression of Cacna1h in SOM+ neurons. Behaviorally, virus-mediated knockdown of Cacna1h in spinal SOM+ neurons reduced the sensitivity to light touch and responsiveness to noxious mechanical stimuli in naïve mice. Furthermore, knockdown of Cacna1h in spinal SOM+ neurons attenuated thermal hyperalgesia and dynamic allodynia in the complete Freund’s adjuvant-induced inflammatory pain model, and reduced both dynamic and static allodynia in a neuropathic pain model of spared nerve injury. Mechanistically, a decrease in the percentage of neurons with Aβ-eEPSCs and Aβ-eAPs in superficial dorsal horn was observed after Cacna1h knockdown in spinal SOM+ neurons. Altogether, our results proved a crucial role of Cav3.2 in spinal SOM+ neurons in mechanosensation under basal conditions and in mechanical allodynia under pathological pain conditions. This work reveals a molecular basis for SOM+ neurons in transmitting mechanical pain and shows a functional role of Cav3.2 in tactile and pain processing at the level of spinal cord in addition to its well-established peripheral role.
Collapse
Affiliation(s)
- Yu-Ru Zhi
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
| | - Feng Cao
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
| | - Xiao-Jing Su
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shu-Wen Gao
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
| | - Hao-Nan Zheng
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Jin-Yan Jiang
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
| | - Li Su
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing, China
| | - Jiao Liu
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing, China
| | - Yun Wang
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yan Zhang
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Ying Zhang,
| | - Ying Zhang
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
- Yan Zhang,
| |
Collapse
|
9
|
Yang N, Liu F, Zhang X, Chen C, Xia Z, Fu S, Wang J, Xu J, Cui S, Zhang Y, Yi M, Wan Y, Li Q, Xu S. A Hybrid Titanium-Softmaterial, High-Strength, Transparent Cranial Window for Transcranial Injection and Neuroimaging. BIOSENSORS 2022; 12:bios12020129. [PMID: 35200389 PMCID: PMC8870569 DOI: 10.3390/bios12020129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 05/04/2023]
Abstract
A transparent and penetrable cranial window is essential for neuroimaging, transcranial injection and comprehensive understanding of cortical functions. For these applications, cranial windows made from glass coverslip, polydimethylsiloxane (PDMS), polymethylmethacrylate, crystal and silicone hydrogel have offered remarkable convenience. However, there is a lack of high-strength, high-transparency, penetrable cranial window with clinical application potential. We engineer high-strength hybrid Titanium-PDMS (Ti-PDMS) cranial windows, which allow large transparent area for in vivo two-photon imaging, and provide a soft window for transcranial injection. Laser scanning and 3D printing techniques are used to match the hybrid cranial window to different skull morphology. A multi-cycle degassing pouring process ensures a good combination of PDMS and Ti frame. Ti-PDMS cranial windows have a high fracture strength matching human skull bone, excellent light transmittance up to 94.4%, and refractive index close to biological tissue. Ti-PDMS cranial windows show excellent bio-compatibility during 21-week implantation in mice. Dye injection shows that the PDMS window has a "self-sealing" to keep liquid from leaking out. Two-photon imaging for brain tissues could be achieved up to 450 µm in z-depth. As a novel brain-computer-interface, this Ti-PDMS device offers an alternative choice for in vivo drug delivery, optical experiments, ultrasonic treatment and electrophysiology recording.
Collapse
Affiliation(s)
- Nana Yang
- Key Laboratory for the Physics & Chemistry of Nanodevices, Department of Electronics, Peking University, Beijing 100871, China; (N.Y.); (J.X.)
| | - Fengyu Liu
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (C.C.); (S.F.); (J.W.); (S.C.); (Y.Z.); (M.Y.); (Y.W.)
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100191, China
- Correspondence: (F.L.); (S.X.)
| | - Xinyue Zhang
- Center of Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China; (X.Z.); (Q.L.)
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
| | - Chenni Chen
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (C.C.); (S.F.); (J.W.); (S.C.); (Y.Z.); (M.Y.); (Y.W.)
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100191, China
| | - Zhiyuan Xia
- Department of Material Science and Engineering, College of Engineering, Peking University, Beijing 100871, China;
| | - Su Fu
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (C.C.); (S.F.); (J.W.); (S.C.); (Y.Z.); (M.Y.); (Y.W.)
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100191, China
| | - Jiaxin Wang
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (C.C.); (S.F.); (J.W.); (S.C.); (Y.Z.); (M.Y.); (Y.W.)
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100191, China
| | - Jingjing Xu
- Key Laboratory for the Physics & Chemistry of Nanodevices, Department of Electronics, Peking University, Beijing 100871, China; (N.Y.); (J.X.)
- School of Microelectronics, Shandong University, Jinan 250100, China
| | - Shuang Cui
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (C.C.); (S.F.); (J.W.); (S.C.); (Y.Z.); (M.Y.); (Y.W.)
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100191, China
| | - Yong Zhang
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (C.C.); (S.F.); (J.W.); (S.C.); (Y.Z.); (M.Y.); (Y.W.)
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100191, China
| | - Ming Yi
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (C.C.); (S.F.); (J.W.); (S.C.); (Y.Z.); (M.Y.); (Y.W.)
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100191, China
| | - You Wan
- Neuroscience Research Institute, Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (C.C.); (S.F.); (J.W.); (S.C.); (Y.Z.); (M.Y.); (Y.W.)
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100191, China
| | - Qing Li
- Center of Digital Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China; (X.Z.); (Q.L.)
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
| | - Shengyong Xu
- Key Laboratory for the Physics & Chemistry of Nanodevices, Department of Electronics, Peking University, Beijing 100871, China; (N.Y.); (J.X.)
- Correspondence: (F.L.); (S.X.)
| |
Collapse
|
10
|
Shin SM, Wang F, Qiu C, Itson-Zoske B, Hogan QH, Yu H. Sigma-1 receptor activity in primary sensory neurons is a critical driver of neuropathic pain. Gene Ther 2022; 29:1-15. [PMID: 32424233 PMCID: PMC7671947 DOI: 10.1038/s41434-020-0157-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/21/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022]
Abstract
The Sigma-1 receptor (σ1R) is highly expressed in the primary sensory neurons (PSNs) that are the critical site of initiation and maintenance of pain following peripheral nerve injury. By immunoblot and immunohistochemistry, we observed increased expression of both σ1R and σ1R-binding immunoglobulin protein (BiP) in the lumbar (L) dorsal root ganglia (DRG) ipsilateral to painful neuropathy induced by spared nerve injury (SNI). To evaluate the therapeutic potential of PSN-targeted σ1R inhibition at a selected segmental level, we designed a recombinant adeno-associated viral (AAV) vector expressing a small hairpin RNA (shRNA) against rat σ1R. Injection of this vector into the L4/L5 DRGs induced downregulation of σ1R in DRG neurons of all size groups, while expression of BiP was not affected. This was accompanied by attenuation of SNI-induced cutaneous mechanical and thermal hypersensitivity. Whole-cell current-clamp recordings of dissociated neurons showed that knockdown of σ1R suppressed neuronal excitability, suggesting that σ1R silencing attenuates pain by reversal of injury-induced neuronal hyperexcitability. These findings support a critical role of σ1R in modulating PSN nociceptive functions, and that the nerve injury-induced elevated σ1R activity in the PSNs can be a significant driver of neuropathic pain. Further understanding the role of PSN-σ1R in pain pathology may open routes to exploit this system for DRG-targeted pain therapy.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA
| | - Fei Wang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, PR China
| | - Chensheng Qiu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266000, PR China
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA.
| |
Collapse
|
11
|
Voltage-dependent Ca V3.2 and Ca V2.2 channels in nociceptive pathways. Pflugers Arch 2022; 474:421-434. [PMID: 35043234 DOI: 10.1007/s00424-022-02666-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 10/19/2022]
Abstract
Noxious stimuli like cold, heat, pH change, tissue damage, and inflammation depolarize a membrane of peripheral endings of specialized nociceptive neurons which eventually results in the generation of an action potential. The electrical signal is carried along a long axon of nociceptive neurons from peripheral organs to soma located in dorsal root ganglions and further to the dorsal horn of the spinal cord where it is transmitted through a chemical synapse and is carried through the spinal thalamic tract into the brain. Two subtypes of voltage-activated calcium play a major role in signal transmission: a low voltage-activated CaV3.2 channel and a high voltage-activated CaV2.2 channel. The CaV3.2 channel contributes mainly to the signal conductance along nociceptive neurons while the principal role of the CaV2.2 channel is in the synaptic transmission at the dorsal horn. Both channels contribute to the signal initiation at peripheral nerve endings. This review summarizes current knowledge about the expression and distribution of these channels in a nociceptive pathway, the regulation of their expression and gating during pain pathology, and their suitability as targets for pharmacological therapy.
Collapse
|
12
|
Gomez K, Vargas-Parada A, Duran P, Sandoval A, Delgado-Lezama R, Khanna R, Felix R. L5-6 Spinal Nerve Ligation-induced Neuropathy Changes the Location and Function of Ca 2+ Channels and Cdk5 and Affects the Compound Action Potential in Adjacent Intact L4 Afferent Fibers. Neuroscience 2021; 471:20-31. [PMID: 34303780 PMCID: PMC8384716 DOI: 10.1016/j.neuroscience.2021.07.013] [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: 04/27/2021] [Revised: 06/30/2021] [Accepted: 07/14/2021] [Indexed: 01/07/2023]
Abstract
Voltage-gated Ca2+ (CaV) channels regulate multiple cell processes, including neurotransmitter release, and have been associated with several pathological conditions, such as neuropathic pain. Cdk5, a neuron-specific kinase, may phosphorylate CaV channels, altering their functional expression. During peripheral nerve injury, upregulation of CaV channels and Cdk5 in the dorsal root ganglia (DRG) and the spinal cord, has been correlated with allodynia. We recently reported an increase in the amplitude of the C component of the compound action potential (cAP) of afferent fibers in animals with allodynia induced by L5-6 spinal nerve ligation (SNL), recorded in the corresponding dorsal roots. This was related to an increase in T-type (CaV3.2) channels generated by Cdk5-mediated phosphorylation. Here, we show that CaV channel functional expression is also altered in the L4 adjacent intact afferent fibers in rats with allodynia induced by L5-6 SNL. Western blot analysis showed that both Cdk5 and CaV3.2 total levels are not increased in the DRG L3-4, but their subcellular distribution changes by concentrating on the neuronal soma. Likewise, the Cdk5 inhibitor olomoucine affected the rapid and the slow C components of the cAP recorded in the dorsal roots. Patch-clamp recordings revealed an increase in T- and N-type currents recorded in the soma of acute isolated L3-4 sensory neurons after L5-6 SNL, which was prevented by olomoucine. These findings suggest changes in CaV channels location and function in L3-4 afferent fibers associated with Cdk5-mediated phosphorylation after L5-6 SNL, which may contribute to nerve injury-induced allodynia.
Collapse
Affiliation(s)
- Kimberly Gomez
- Department of Physiology, Biophysics and Neuroscience, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Alberto Vargas-Parada
- Department of Physiology, Biophysics and Neuroscience, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Paz Duran
- Department of Cell Biology, Cinvestav, Mexico City, Mexico
| | - Alejandro Sandoval
- School of Medicine FES Iztacala, National Autonomous University of Mexico (UNAM), Tlalnepantla, Mexico
| | - Rodolfo Delgado-Lezama
- Department of Physiology, Biophysics and Neuroscience, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Ricardo Felix
- Department of Cell Biology, Cinvestav, Mexico City, Mexico.
| |
Collapse
|
13
|
Targeting T-type/CaV3.2 channels for chronic pain. Transl Res 2021; 234:20-30. [PMID: 33422652 PMCID: PMC8217081 DOI: 10.1016/j.trsl.2021.01.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/31/2020] [Accepted: 01/04/2021] [Indexed: 01/09/2023]
Abstract
T-type calcium channels regulate neuronal excitability and are important contributors of pain processing. CaV3.2 channels are the major isoform expressed in nonpeptidergic and peptidergic nociceptive neurons and are emerging as promising targets for pain treatment. Numerous studies have shown that CaV3.2 expression and/or activity are significantly increased in spinal dorsal horn and in dorsal root ganglia neurons in different inflammatory and neuropathic pain models. Pharmacological campaigns to inhibit the functional expression of CaV3.2 for treatment of pain have focused on the development of direct channel blockers, but none have produced lead candidates. Targeting the proteins that regulate the trafficking or transcription, and the ones that modify the channels via post-translational modifications are alternative means to regulate expression and function of CaV3.2 channels and hence to develop new drugs to control pain. Here we synthesize data supporting a role for CaV3.2 in numerous pain modalities and then discuss emerging opportunities for the indirect targeting of CaV3.2 channels.
Collapse
|
14
|
Boscia F, Elkjaer ML, Illes Z, Kukley M. Altered Expression of Ion Channels in White Matter Lesions of Progressive Multiple Sclerosis: What Do We Know About Their Function? Front Cell Neurosci 2021; 15:685703. [PMID: 34276310 PMCID: PMC8282214 DOI: 10.3389/fncel.2021.685703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/23/2021] [Indexed: 12/19/2022] Open
Abstract
Despite significant advances in our understanding of the pathophysiology of multiple sclerosis (MS), knowledge about contribution of individual ion channels to axonal impairment and remyelination failure in progressive MS remains incomplete. Ion channel families play a fundamental role in maintaining white matter (WM) integrity and in regulating WM activities in axons, interstitial neurons, glia, and vascular cells. Recently, transcriptomic studies have considerably increased insight into the gene expression changes that occur in diverse WM lesions and the gene expression fingerprint of specific WM cells associated with secondary progressive MS. Here, we review the ion channel genes encoding K+, Ca2+, Na+, and Cl- channels; ryanodine receptors; TRP channels; and others that are significantly and uniquely dysregulated in active, chronic active, inactive, remyelinating WM lesions, and normal-appearing WM of secondary progressive MS brain, based on recently published bulk and single-nuclei RNA-sequencing datasets. We discuss the current state of knowledge about the corresponding ion channels and their implication in the MS brain or in experimental models of MS. This comprehensive review suggests that the intense upregulation of voltage-gated Na+ channel genes in WM lesions with ongoing tissue damage may reflect the imbalance of Na+ homeostasis that is observed in progressive MS brain, while the upregulation of a large number of voltage-gated K+ channel genes may be linked to a protective response to limit neuronal excitability. In addition, the altered chloride homeostasis, revealed by the significant downregulation of voltage-gated Cl- channels in MS lesions, may contribute to an altered inhibitory neurotransmission and increased excitability.
Collapse
Affiliation(s)
- Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Maria Louise Elkjaer
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Zsolt Illes
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Maria Kukley
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
15
|
Whole Genomic DNA Methylation Profiling of CpG Sites in Promoter Regions of Dorsal Root Ganglion in Diabetic Neuropathic Pain Mice. J Mol Neurosci 2021; 71:2558-2565. [PMID: 33950354 DOI: 10.1007/s12031-021-01847-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
DNA methylation and demethylation play an important role in neuropathic pain. In general, DNA methylation of CpG sites in the promoter region impedes gene expression, whereas DNA demethylation contributes to gene expression. Here, we evaluated the methylation status of CpG sites in genomic DNA promoter regions in dorsal root ganglions (DRGs) of diabetic neuropathic pain (DNP) mice. In our research, streptozotocin (STZ) was intraperitoneally injected into mice to construct DNP models. The DNP mice showed higher fasting blood glucose (above 11.1 mmol/L), lower body weight, and mechanical allodynia than control mice. Whole-genome bisulfite sequencing (WGBS) revealed an altered methylation pattern in CpG sites in the DNA promoter regions in DRGs of DNP mice. The results showed 376 promoter regions with hypermethylated CpG sites and 336 promoter regions with hypomethylated CpG sites. In addition, our data indicated that altered DNA methylation occurs primarily on CpG sites in DNA promoter regions. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that differentially methylated CpG sites annotated genes were involved in activities of the nervous and sensory systems. Enrichment analysis indicated that genes in these pathways contributed to diabetes or pain. In conclusion, our study enriched the role of DNA methylation in DNP.
Collapse
|
16
|
A modulator of the low-voltage-activated T-type calcium channel that reverses HIV glycoprotein 120-, paclitaxel-, and spinal nerve ligation-induced peripheral neuropathies. Pain 2021; 161:2551-2570. [PMID: 32541387 DOI: 10.1097/j.pain.0000000000001955] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The voltage-gated calcium channels CaV3.1-3.3 constitute the T-type subfamily, whose dysfunctions are associated with epilepsy, psychiatric disorders, and chronic pain. The unique properties of low-voltage-activation, faster inactivation, and slower deactivation of these channels support their role in modulation of cellular excitability and low-threshold firing. Thus, selective T-type calcium channel antagonists are highly sought after. Here, we explored Ugi-azide multicomponent reaction products to identify compounds targeting T-type calcium channel. Of the 46 compounds tested, an analog of benzimidazolonepiperidine-5bk (1-{1-[(R)-{1-[(1S)-1-phenylethyl]-1H-1,2,3,4-tetrazol-5-yl}(thiophen-3-yl)methyl]piperidin-4-yl}-2,3-dihydro-1H-1,3-benzodiazol-2-one) modulated depolarization-induced calcium influx in rat sensory neurons. Modulation of T-type calcium channels by 5bk was further confirmed in whole-cell patch clamp assays in dorsal root ganglion (DRG) neurons, where pharmacological isolation of T-type currents led to a time- and concentration-dependent regulation with a low micromolar IC50. Lack of an acute effect of 5bk argues against a direct action on T-type channels. Genetic knockdown revealed CaV3.2 to be the isoform preferentially modulated by 5bk. High voltage-gated calcium, as well as tetrodotoxin-sensitive and -resistant sodium, channels were unaffected by 5bk. 5bk inhibited spontaneous excitatory postsynaptic currents and depolarization-evoked release of calcitonin gene-related peptide from lumbar spinal cord slices. Notably, 5bk did not bind human mu, delta, or kappa opioid receptors. 5bk reversed mechanical allodynia in rat models of HIV-associated neuropathy, chemotherapy-induced peripheral neuropathy, and spinal nerve ligation-induced neuropathy, without effects on locomotion or anxiety. Thus, 5bk represents a novel T-type modulator that could be used to develop nonaddictive pain therapeutics.
Collapse
|
17
|
Tran EL, Crawford LK. Revisiting PNS Plasticity: How Uninjured Sensory Afferents Promote Neuropathic Pain. Front Cell Neurosci 2020; 14:612982. [PMID: 33362476 PMCID: PMC7759741 DOI: 10.3389/fncel.2020.612982] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/12/2020] [Indexed: 11/13/2022] Open
Abstract
Despite the widespread study of how injured nerves contribute to chronic pain, there are still major gaps in our understanding of pain mechanisms. This is particularly true of pain resulting from nerve injury, or neuropathic pain, wherein tactile or thermal stimuli cause painful responses that are particularly difficult to treat with existing therapies. Curiously, this stimulus-driven pain relies upon intact, uninjured sensory neurons that transmit the signals that are ultimately sensed as painful. Studies that interrogate uninjured neurons in search of cell-specific mechanisms have shown that nerve injury alters intact, uninjured neurons resulting in an activity that drives stimulus-evoked pain. This review of neuropathic pain mechanisms summarizes cell-type-specific pathology of uninjured sensory neurons and the sensory ganglia that house their cell bodies. Uninjured neurons have demonstrated a wide range of molecular and neurophysiologic changes, many of which are distinct from those detected in injured neurons. These intriguing findings include expression of pain-associated molecules, neurophysiological changes that underlie increased excitability, and evidence that intercellular signaling within sensory ganglia alters uninjured neurons. In addition to well-supported findings, this review also discusses potential mechanisms that remain poorly understood in the context of nerve injury. This review highlights key questions that will advance our understanding of the plasticity of sensory neuron subpopulations and clarify the role of uninjured neurons in developing anti-pain therapies.
Collapse
Affiliation(s)
- Emily L Tran
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, WI, United States
| | - LaTasha K Crawford
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, WI, United States
| |
Collapse
|
18
|
Fight fire with fire: Neurobiology of capsaicin-induced analgesia for chronic pain. Pharmacol Ther 2020; 220:107743. [PMID: 33181192 DOI: 10.1016/j.pharmthera.2020.107743] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/04/2020] [Indexed: 12/12/2022]
Abstract
Capsaicin, the pungent ingredient in chili peppers, produces intense burning pain in humans. Capsaicin selectively activates the transient receptor potential vanilloid 1 (TRPV1), which is enriched in nociceptive primary afferents, and underpins the mechanism for capsaicin-induced burning pain. Paradoxically, capsaicin has long been used as an analgesic. The development of topical patches and injectable formulations containing capsaicin has led to application in clinical settings to treat chronic pain conditions, such as neuropathic pain and the potential to treat osteoarthritis. More detailed determination of the neurobiological mechanisms of capsaicin-induced analgesia should provide the logical rationale for capsaicin therapy and help to overcome the treatment's limitations, which include individual differences in treatment outcome and procedural discomfort. Low concentrations of capsaicin induce short-term defunctionalization of nociceptor terminals. This phenomenon is reversible within hours and, hence, likely does not account for the clinical benefit. By contrast, high concentrations of capsaicin lead to long-term defunctionalization mediated by the ablation of TRPV1-expressing afferent terminals, resulting in long-lasting analgesia persisting for several months. Recent studies have shown that capsaicin-induced Ca2+/calpain-mediated ablation of axonal terminals is necessary to produce long-lasting analgesia in a mouse model of neuropathic pain. In combination with calpain, axonal mitochondrial dysfunction and microtubule disorganization may also contribute to the longer-term effects of capsaicin. The analgesic effects subside over time in association with the regeneration of the ablated afferent terminals. Further determination of the neurobiological mechanisms of capsaicin-induced analgesia should lead to more efficacious non-opioidergic analgesic options with fewer adverse side effects.
Collapse
|
19
|
Contribution of T-Type Calcium Channels to Spinal Cord Injury-Induced Hyperexcitability of Nociceptors. J Neurosci 2020; 40:7229-7240. [PMID: 32839232 DOI: 10.1523/jneurosci.0517-20.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 07/22/2020] [Accepted: 07/30/2020] [Indexed: 01/24/2023] Open
Abstract
A hyperexcitable state and spontaneous activity of nociceptors have been suggested to play a critical role in the development of chronic neuropathic pain following spinal cord injury (SCI). In male rats, we employed the action potential-clamp technique to determine the underlying ionic mechanisms responsible for driving SCI-nociceptors to a hyperexcitable state and for triggering their spontaneous activity. We found that the increased activity of low voltage activated T-type calcium channels induced by the injury sustains the bulk (∼60-70%) of the inward current active at subthreshold voltages during the interspike interval in SCI-nociceptors, with a modest contribution (∼10-15%) from tetrodotoxin (TTX)-sensitive and TTX-resistant sodium channels and hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. In current-clamp recordings, inhibition of T-type calcium channels with 1 μm TTA-P2 reduced both the spontaneous and the evoked firing in response to current injections in SCI-nociceptors to a level similar to sham-nociceptors. Electrophysiology in vitro was then combined with the conditioned place preference (CPP) paradigm to determine the relationship between the increased activity of T-type channels in SCI-nociceptors and chronic neuropathic pain following SCI. The size of the interspike T-type calcium current recorded from nociceptors isolated from SCI rats showing TTA-P2-induced CPP (responders) was ∼6 fold greater than the interspike T-type calcium current recorded from nociceptors isolated from SCI rats without TTA-P2-induced CPP (non-responders). Taken together, our data suggest that the increased activity of T-type calcium channels induced by the injury plays a primary role in driving SCI-nociceptors to a hyperexcitable state and contributes to chronic neuropathic pain following SCI.SIGNIFICANCE STATEMENT Chronic neuropathic pain is a major comorbidity of spinal cord injury (SCI), affecting up to 70-80% of patients. Anticonvulsant and tricyclic antidepressant drugs are first line analgesics used to treat SCI-induced neuropathic pain, but their efficacy is very limited. A hyperexcitable state and spontaneous activity of SCI-nociceptors have been proposed as a possible underlying cause for the development of chronic neuropathic pain following SCI. Here, we show that the increased activity of T-type calcium channels induced by the injury plays a major role in driving SCI-nociceptors to a hyperexcitable state and for promoting their spontaneous activity, suggesting that T-type calcium channels may represent a pharmacological target to treat SCI-induced neuropathic pain.
Collapse
|
20
|
Huo R, Han SP, Liu FY, Shou XJ, Liu LY, Song TJ, Zhai FJ, Zhang R, Xing GG, Han JS. Responses of Primary Afferent Fibers to Acupuncture-Like Peripheral Stimulation at Different Frequencies: Characterization by Single-Unit Recording in Rats. Neurosci Bull 2020; 36:907-918. [PMID: 32394277 PMCID: PMC7410905 DOI: 10.1007/s12264-020-00509-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 01/18/2020] [Indexed: 10/24/2022] Open
Abstract
The pain-relieving effect of acupuncture is known to involve primary afferent nerves (PANs) via their roles in signal transmission to the CNS. Using single-unit recording in rats, we characterized the generation and transmission of electrical signals in Aβ and Aδ fibers induced by acupuncture-like stimuli. Acupuncture-like signals were elicited in PANs using three techniques: manual acupuncture (MAc), emulated acupuncture (EAc), and electro-acupuncture (EA)-like peripheral electrical stimulation (PES). The discharges evoked by MAc and EAc were mostly in a burst pattern with average intra-burst and inter-burst firing rates of 90 Hz and 2 Hz, respectively. The frequency of discharges in PANs was correlated with the frequency of PES. The highest discharge frequency was 246 Hz in Aβ fibers and 180 Hz in Aδ fibers. Therefore, EA in a dense-disperse mode (at alternating frequency between 2 Hz and 15 Hz or between 2 Hz and 100 Hz) best mimics MAc. Frequencies of EA output >250 Hz appear to be obsolete for pain relief.
Collapse
Affiliation(s)
- Ran Huo
- Neuroscience Research Institute, Peking University, Beijing, 100191, China
- Department of Neurobiology, Peking University School of Basic Medical Sciences, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191, China
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China
| | - Song-Ping Han
- Neuroscience Research Institute, Peking University, Beijing, 100191, China
- Department of Neurobiology, Peking University School of Basic Medical Sciences, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191, China
| | - Feng-Yu Liu
- Neuroscience Research Institute, Peking University, Beijing, 100191, China
- Department of Neurobiology, Peking University School of Basic Medical Sciences, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191, China
| | - Xiao-Jing Shou
- Neuroscience Research Institute, Peking University, Beijing, 100191, China
- Department of Neurobiology, Peking University School of Basic Medical Sciences, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191, China
| | - Ling-Yu Liu
- Neuroscience Research Institute, Peking University, Beijing, 100191, China
- Department of Neurobiology, Peking University School of Basic Medical Sciences, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191, China
| | - Tian-Jia Song
- Neuroscience Research Institute, Peking University, Beijing, 100191, China
- Department of Neurobiology, Peking University School of Basic Medical Sciences, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191, China
| | - Fu-Jun Zhai
- Neuroscience Research Institute, Peking University, Beijing, 100191, China
- Department of Neurobiology, Peking University School of Basic Medical Sciences, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191, China
| | - Rong Zhang
- Neuroscience Research Institute, Peking University, Beijing, 100191, China
- Department of Neurobiology, Peking University School of Basic Medical Sciences, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191, China
- Department of Integration of Chinese and Western Medicine, Peking University School of Basic Medical Sciences, Beijing, 100191, China
| | - Guo-Gang Xing
- Neuroscience Research Institute, Peking University, Beijing, 100191, China.
- Department of Neurobiology, Peking University School of Basic Medical Sciences, Beijing, 100191, China.
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191, China.
- Department of Integration of Chinese and Western Medicine, Peking University School of Basic Medical Sciences, Beijing, 100191, China.
| | - Ji-Sheng Han
- Neuroscience Research Institute, Peking University, Beijing, 100191, China.
- Department of Neurobiology, Peking University School of Basic Medical Sciences, Beijing, 100191, China.
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191, China.
| |
Collapse
|
21
|
Abstract
Neuropathic pain caused by a lesion or disease of the somatosensory nervous system is a common chronic pain condition with major impact on quality of life. Examples include trigeminal neuralgia, painful polyneuropathy, postherpetic neuralgia, and central poststroke pain. Most patients complain of an ongoing or intermittent spontaneous pain of, for example, burning, pricking, squeezing quality, which may be accompanied by evoked pain, particular to light touch and cold. Ectopic activity in, for example, nerve-end neuroma, compressed nerves or nerve roots, dorsal root ganglia, and the thalamus may in different conditions underlie the spontaneous pain. Evoked pain may spread to neighboring areas, and the underlying pathophysiology involves peripheral and central sensitization. Maladaptive structural changes and a number of cell-cell interactions and molecular signaling underlie the sensitization of nociceptive pathways. These include alteration in ion channels, activation of immune cells, glial-derived mediators, and epigenetic regulation. The major classes of therapeutics include drugs acting on α2δ subunits of calcium channels, sodium channels, and descending modulatory inhibitory pathways.
Collapse
Affiliation(s)
- Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Rohini Kuner
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
22
|
Cardoso FC. Multi-targeting sodium and calcium channels using venom peptides for the treatment of complex ion channels-related diseases. Biochem Pharmacol 2020; 181:114107. [PMID: 32579958 DOI: 10.1016/j.bcp.2020.114107] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/13/2020] [Accepted: 06/17/2020] [Indexed: 02/08/2023]
Abstract
Venom peptides are amongst the most exquisite group of bioactive molecules able to alter the normal physiology of organisms. These bioactive peptides penetrate tissues and blood vessels to encounter a number of receptors and ion channels to which they bind with high affinity and execute modulatory activities. Arachnid is the most diverse class of venomous animals often rich in peptides modulating voltage-gated sodium (NaV), calcium (CaV), and potassium (KV) channels. Spider venoms, in particular, contain potent and selective peptides targeting these channels, with a few displaying interesting multi-target properties for NaV and CaV channels underlying disease mechanisms such as in neuropathic pain, motor neuron disease and cancer. The elucidation of the pharmacology and structure-function properties of these venom peptides are invaluable for the development of effective drugs targeting NaV and CaV channels. This perspective discusses spider venom peptides displaying multi-target properties to modulate NaV and CaV channels in regard to their pharmacological features, structure-function relationships and potential to become the next generation of effective drugs to treat neurological disorders and other multi-ion channels related diseases.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd., St Lucia, QLD AU 4072, Australia
| |
Collapse
|
23
|
Jeevakumar V, Al Sardar AK, Mohamed F, Smithhart CM, Price T, Dussor G. IL-6 induced upregulation of T-type Ca 2+ currents and sensitization of DRG nociceptors is attenuated by MNK inhibition. J Neurophysiol 2020; 124:274-283. [PMID: 32519575 DOI: 10.1152/jn.00188.2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Phosphorylation of the 5' cap-binding protein eIF4E by MAPK-interacting kinases (MNK1/2) is important for nociceptor sensitization and the development of chronic pain. IL-6-induced dorsal root ganglion (DRG) nociceptor excitability is attenuated in mice lacking eIF4E phosphorylation, in MNK1/2-/- mice, and by the nonselective MNK1/2 inhibitor cercosporamide. Here, we sought to better understand the neurophysiological mechanisms underlying how IL-6 causes nociceptor excitability via MNK-eIF4E signaling using the highly selective MNK inhibitor eFT508. DRG neurons were cultured from male and female ICR mice, 4-7 wk old. DRG cultures were treated with vehicle, IL-6, eFT508 (pretreat) followed by IL-6, or eFT508 alone. Whole cell patch-clamp recordings were done on small-diameter neurons (20-30 pF) to measure membrane excitability in response to ramp depolarization. IL-6 treatment (1 h) resulted in increased action potential firing compared with vehicle at all ramp intensities, an effect that was blocked by pretreatment with eFT508. Basic membrane properties, including resting membrane potential, input resistance, and rheobase, were similar across groups. Latency to the first action potential in the ramp protocol was lower in the IL-6 group and rescued by eFT508 pretreatment. We also found that the amplitudes of T-type voltage-gated calcium channels (VGCCs) were increased in the DRG following IL-6 treatment, but not in the eFT508 cotreatment group. Our findings are consistent with a model wherein MNK-eIF4E signaling controls the translation of signaling factors that regulate T-type VGCCs in response to IL-6 treatment. Inhibition of MNK with eFT508 disrupts these events, thereby preventing nociceptor hyperexcitability.NEW & NOTEWORTHY In this study, we show that the MNK inhibitor and anti-tumor agent eFT508 (tomivosertib) is effective in attenuating IL-6 induced sensitization of dorsal root ganglion (DRG) nociceptors. Pretreatment with eFT508 in DRG cultures from mice helps mitigate the development of hyperexcitability in response to IL-6. Furthermore, our data reveal that the upregulation of T-type voltage-gated calcium channels following IL-6 application can be blocked by eFT508, implicating the MNK-eIF4E signaling pathway in membrane trafficking of ion channels.
Collapse
Affiliation(s)
- Vivek Jeevakumar
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas
| | - Aysha Khalid Al Sardar
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas
| | - Farah Mohamed
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas
| | - Clay Matthew Smithhart
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas
| | - Theodore Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas
| |
Collapse
|
24
|
Shin SM, Cai Y, Itson-Zoske B, Qiu C, Hao X, Xiang H, Hogan QH, Yu H. Enhanced T-type calcium channel 3.2 activity in sensory neurons contributes to neuropathic-like pain of monosodium iodoacetate-induced knee osteoarthritis. Mol Pain 2020; 16:1744806920963807. [PMID: 33054557 PMCID: PMC7570798 DOI: 10.1177/1744806920963807] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
The monosodium iodoacetate knee osteoarthritis model has been widely used for the evaluation of osteoarthritis pain, but the pathogenesis of associated chronic pain is not fully understood. The T-type calcium channel 3.2 (CaV3.2) is abundantly expressed in the primary sensory neurons, in which it regulates neuronal excitability at both the somata and peripheral terminals and facilitates spontaneous neurotransmitter release at the spinal terminals. In this study, we investigated the involvement of primary sensory neuron-CaV3.2 activation in monosodium iodoacetate osteoarthritis pain. Knee joint osteoarthritis pain was induced by intra-articular injection of monosodium iodoacetate (2 mg) in rats, and sensory behavior was evaluated for 35 days. At that time, knee joint structural histology, primary sensory neuron injury, and inflammatory gliosis in lumbar dorsal root ganglia, and spinal dorsal horn were examined. Primary sensory neuron-T-type calcium channel current by patch-clamp recording and CaV3.2 expression by immunohistochemistry and immunoblots were determined. In a subset of animals, pain relief by CaV3.2 inhibition after delivery of CaV3.2 inhibitor TTA-P2 into sciatic nerve was investigated. Knee injection of monosodium iodoacetate resulted in osteoarthritis histopathology, weight-bearing asymmetry, sensory hypersensitivity of the ipsilateral hindpaw, and inflammatory gliosis in the ipsilateral dorsal root ganglia, sciatic nerve, and spinal dorsal horn. Neuronal injury marker ATF-3 was extensively upregulated in primary sensory neurons, suggesting that neuronal damage was beyond merely knee-innervating primary sensory neurons. T-type current in dissociated primary sensory neurons from lumbar dorsal root ganglia of monosodium iodoacetate rats was significantly increased, and CaV3.2 protein levels in the dorsal root ganglia and spinal dorsal horn ipsilateral to monosodium iodoacetate by immunoblots were significantly increased, compared to controls. Perineural application of TTA-P2 into the ipsilateral sciatic nerve alleviated mechanical hypersensitivity and weight-bearing asymmetry in monosodium iodoacetate osteoarthritis rats. Overall, our findings demonstrate an elevated CaV3.2 expression and enhanced function of primary sensory neuron-T channels in the monosodium iodoacetate osteoarthritis pain. Further study is needed to delineate the importance of dysfunctional primary sensory neuron-CaV3.2 in osteoarthritis pain.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
| | - Yongsong Cai
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Xi’an Honghui Hospital, Xi’an, Shaanxi, PR China
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Chensheng Qiu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - Xu Hao
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - Hongfei Xiang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
| |
Collapse
|
25
|
Sharma J, Maslov LN, Singh N, Jaggi AS. Pain attenuating actions of vincristinet-preconditioning in chemotherapeutic agent-induced neuropathic pain: key involvement of T-type calcium channels. Fundam Clin Pharmacol 2019; 34:336-344. [PMID: 31797451 DOI: 10.1111/fcp.12519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 11/02/2019] [Accepted: 11/04/2019] [Indexed: 12/17/2022]
Abstract
Preconditioning is a well-documented strategy that induces hepatic protection, renal protection, cardioprotection, and neuroprotection but its mechanism still remains to be elucidated. Hence, the present study investigated the protective mechanism underlying pain attenuating effects of vincristine-preconditioning in chemotherapeutic agent-induced neuropathic pain. Neuropathic pain was induced by administration of vincristine (50 µg/kg, i.p.) for 10 days in rats. Vincristine-preconditioning was induced by administration of vincristine (2, 5, and 10 µg/kg, i.p) for 5 days before administration of pain-inducing dose of vincristine (50 µg/kg, i.p.). Vincristine-preconditioning (10 µg/kg, i.p) for 5 days significantly reduced vincristine (50 µg/kg, i.p.) induced pain-related behaviors including paw cold allodynia, mechanical hyperalgesia, and heat hyperalgesia. However, vincristine (2 and 5 µg/kg, i.p) did not significantly ameliorate the vincristine (50 µg/kg, i.p.) induced neuropathic pain in rats. Furthermore, to explore the involvement of calcium channels in pain attenuating mechanism of vincristine-preconditioning, T-type calcium channel blocker, ethosuximide (100 and 200 mg/kg, i.p.) and L-type calcium channel blocker, amlodipine (5 and 10 mg/kg, i.p.) were used. Pretreatment with T-type calcium channel blocker, ethosuximide significantly abolished vincristine-preconditioning-induced protective effect. However, pretreatment with L-type calcium channel blocker, amlodipine did not alter vincristine-preconditioning-induced pain-related behaviors. This indicates that vincristine-preconditioning has protective effect on pain-related parameters due to opening of calcium channels, particularly T-type calcium channels that lead to entry of small magnitude of intracellular calcium through these channels and prevent the deleterious effects of high-dose vincristine.
Collapse
Affiliation(s)
- Jasmine Sharma
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, 147002, Patiala, India
| | - Leonid N Maslov
- Laboratory of Experimental Cardiology, Institute of Cardiology, Kyevskaya 111, 634012, Tomsk, Russia
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, 147002, Patiala, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, 147002, Patiala, India
| |
Collapse
|
26
|
Cdk5-Dependent Phosphorylation of Ca V3.2 T-Type Channels: Possible Role in Nerve Ligation-Induced Neuropathic Allodynia and the Compound Action Potential in Primary Afferent C Fibers. J Neurosci 2019; 40:283-296. [PMID: 31744861 DOI: 10.1523/jneurosci.0181-19.2019] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 11/05/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Voltage-gated T-type Ca2+ (CaV3) channels regulate diverse physiological events, including neuronal excitability, and have been linked to several pathological conditions such as absence epilepsy, cardiovascular diseases, and neuropathic pain. It is also acknowledged that calcium/calmodulin-dependent protein kinase II and protein kinases A and C regulate the activity of T-type channels. Interestingly, peripheral nerve injury induces tactile allodynia and upregulates CaV3.2 channels and cyclin-dependent kinase 5 (Cdk5) in dorsal root ganglia (DRG) and spinal dorsal horn. Here, we report that recombinant CaV3.2 channels expressed in HEK293 cells are regulatory targets of Cdk5. Site-directed mutagenesis showed that the relevant sites for this regulation are residues S561 and S1987. We also found that Cdk5 may regulate CaV3.2 channel functional expression in rats with mechanical allodynia induced by spinal nerve ligation (SNL). Consequently, the Cdk5 inhibitor olomoucine affected the compound action potential recorded in the spinal nerves, as well as the paw withdrawal threshold. Likewise, Cdk5 expression was upregulated after SNL in the DRG. These findings unveil a novel mechanism for how phosphorylation may regulate CaV3.2 channels and suggest that increased channel activity by Cdk5-mediated phosphorylation after SNL contributes nerve injury-induced tactile allodynia.SIGNIFICANCE STATEMENT Neuropathic pain is a current public health challenge. It can develop as a result of injury or nerve illness. It is acknowledged that the expression of various ion channels can be altered in neuropathic pain, including T-type Ca2+ channels that are expressed in sensory neurons, where they play a role in the regulation of cellular excitability. The present work shows that the exacerbated expression of Cdk5 in a preclinical model of neuropathic pain increases the functional expression of CaV3.2 channels. This finding is relevant for the understanding of the molecular pathophysiology of the disease. Additionally, this work may have a substantial translational impact, since it describes a novel molecular pathway that could represent an interesting therapeutic alternative for neuropathic pain.
Collapse
|
27
|
Liu Q, Chen W, Fan X, Wang J, Fu S, Cui S, Liao F, Cai J, Wang X, Huang Y, Su L, Zhong L, Yi M, Liu F, Wan Y. Upregulation of interleukin-6 on Cav3.2 T-type calcium channels in dorsal root ganglion neurons contributes to neuropathic pain in rats with spinal nerve ligation. Exp Neurol 2019; 317:226-243. [DOI: 10.1016/j.expneurol.2019.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/22/2019] [Accepted: 03/09/2019] [Indexed: 10/27/2022]
|
28
|
Upregulation of Ca v3.2 T-type calcium channels in adjacent intact L4 dorsal root ganglion neurons in neuropathic pain rats with L5 spinal nerve ligation. Neurosci Res 2018; 142:30-37. [PMID: 29684385 DOI: 10.1016/j.neures.2018.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 02/07/2023]
Abstract
Besides the injured peripheral dorsal root ganglion (DRG) neurons, the adjacent intact DRG neurons also have important roles in neuropathic pain. Ion channels including Cav3.2 T-type calcium channel in the DRG neurons are important in the development of neuropathic pain. In the present study, we aimed to examine the expression of Cav3.2 T-type calcium channels in the intact DRG neurons in neuropathic pain. A neuropathic pain model of rat with lumbar 5 (L5) spinal nerve ligation (SNL) was established, in which the L4 DRG was separated from the axotomized L5 DRG, and the molecular, morphological and electrophysiological changes of Cav3.2 T-type calcium channels in L4 DRG neurons were investigated. Western blotting showed that total and membrane protein levels of Cav3.2 in L4 DRG neurons increased, and voltage-dependent patch clamp recordings revealed an increased T-type current density with a curve shift to the left in steady-state activation in the acutely isolated L4 DRG neurons in neuropathic pain rats. Immunofluorescent staining further showed that the membrane expression of Cav3.2 increased in CGRP-, IB4-positive small neurons and NF200-positive large ones. In conclusion, the membrane expression and the function of Cav3.2 T-type calcium channels are increased in the intact L4 DRG neurons in neuropathic pain rats with peripheral nerve injury like SNL.
Collapse
|