1
|
Jiang BC, Ling YJ, Xu ML, Gu J, Wu XB, Sha WL, Tian T, Bai XH, Li N, Jiang CY, Chen O, Ma LJ, Zhang ZJ, Qin YB, Zhu M, Yuan HJ, Wu LJ, Ji RR, Gao YJ. Follistatin drives neuropathic pain in mice through IGF1R signaling in nociceptive neurons. Sci Transl Med 2024; 16:eadi1564. [PMID: 39413164 DOI: 10.1126/scitranslmed.adi1564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/10/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
Neuropathic pain is a debilitating chronic condition that lacks effective treatment. The role of cytokine- and chemokine-mediated neuroinflammation in its pathogenesis has been well documented. Follistatin (FST) is a secreted protein known to antagonize the biological activity of cytokines in the transforming growth factor-β (TGF-β) superfamily. The involvement of FST in neuropathic pain and the underlying mechanism remain largely unknown. Here, we report that FST was up-regulated in A-fiber sensory neurons after spinal nerve ligation (SNL) in mice. Inhibition or deletion of FST alleviated neuropathic pain and reduced the nociceptive neuron hyperexcitability induced by SNL. Conversely, intrathecal or intraplantar injection of recombinant FST, or overexpression of FST in the dorsal root ganglion (DRG) neurons, induced pain hypersensitivity. Furthermore, exogenous FST increased neuronal excitability in nociceptive neurons. The biolayer interferometry (BLI) assay and coimmunoprecipitation (co-IP) demonstrated direct binding of FST to the insulin-like growth factor-1 receptor (IGF1R), and IGF1R inhibition reduced FST-induced activation of extracellular signal-regulated kinase (ERK) and protein kinase B (AKT), as well as neuronal hyperexcitability. Further co-IP analysis revealed that the N-terminal domain of FST exhibits the highest affinity for IGF1R, and blocking this interaction with a peptide derived from FST attenuated Nav1.7-mediated neuronal hyperexcitability and neuropathic pain after SNL. In addition, FST enhanced neuronal excitability in human DRG neurons through IGF1R. Collectively, our findings suggest that FST, released from A-fiber neurons, enhances Nav1.7-mediated hyperexcitability of nociceptive neurons by binding to IGF1R, making it a potential target for neuropathic pain treatment.
Collapse
Affiliation(s)
- Bao-Chun Jiang
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Yue-Juan Ling
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Meng-Lin Xu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Jun Gu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Xiao-Bo Wu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Wei-Lin Sha
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Tian Tian
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Xue-Hui Bai
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Nan Li
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong 518052, China
| | - Chang-Yu Jiang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong 518052, China
| | - Ouyang Chen
- Center for Translational Pain Medicine, Departments of Anesthesiology, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ling-Jie Ma
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Zhi-Jun Zhang
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Yi-Bin Qin
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| | - Meixuan Zhu
- Department of Psychology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hong-Jie Yuan
- Department of Pain Management, Nantong Hospital of Traditional Chinese Medicine, Jiangsu 226001, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Departments of Anesthesiology, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yong-Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Department of Pain Management of the Affiliated Hospital, Nantong University, Jiangsu 226019, China
| |
Collapse
|
2
|
Santi MD, Zhang M, Asam K, Yu G, Dong PM, Sheehan DH, Aouizerat BE, Thomas CM, Viet CT, Ye Y. Perineural Invasion Is Associated With Function-evoked Pain and Altered Extracellular Matrix in Patients With Head and Neck Squamous Cell Carcinoma. THE JOURNAL OF PAIN 2024; 25:104615. [PMID: 38936749 DOI: 10.1016/j.jpain.2024.104615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/01/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is painful, and perineural invasion (PNI) has been associated with the worst pain. Pain due to HNSCC is diverse and may vary based on clinicopathological factors. This study aims to characterize different pain patterns linked with PNI, its influence on daily functioning, and gain insights into molecular changes and pathways associated with PNI-related pain in HNSCC patients. We conducted a cross-sectional study across 3 medical centers (n = 114), assessing pain phenotypes and their impact on daily functioning using 2 self-reported pain questionnaires, given to patients prior to their cancer surgery. Furthermore, we conducted RNA-seq analysis utilizing the The Cancer Genome Atlas dataset of HNSCC tumor from patients (n = 192) to identify genes relevant to both PNI and pain. Upon adjusting for demographic and clinicopathological variables using linear regression models, we found that PNI independently predicted function-evoked pain according to the University of Calfornia San Francisco Oral Cancer Pain Questionnaire, as well as the worst pain intensity reported in the Brief Pain Inventory. Distinct pain patterns were observed to be associated with daily activities in varying manners. Our molecular analyses revealed significant disruptions in pathways associated with the extracellular matrix structure and organization. The top differentially expressed genes linked to the extracellular matrix are implicated in cancer development, pain, and neurodegenerative diseases. Our data underscore the importance of properly categorizing pain phenotypes in future studies aiming to uncover mechanistic underpinnings of pain. Additionally, we have compiled a list of genes of interest that could serve as targets for both cancer and cancer pain management. PERSPECTIVE: PNI independently predicts function-evoked pain. Different pain phenotypes affect daily activities differently. We identified a list of candidate genes involved in the extracellular matrix structure and function that can be targeted for both cancer and cancer pain control.
Collapse
Affiliation(s)
- Maria D Santi
- Translational Research Center, College of Dentistry, New York University, New York, New York; Department of Molecular Pathobiology, Pain Research Center, College of Dentistry, New York University, New York, New York
| | - Morgan Zhang
- Translational Research Center, College of Dentistry, New York University, New York, New York; Department of Molecular Pathobiology, Pain Research Center, College of Dentistry, New York University, New York, New York
| | - Kesava Asam
- Translational Research Center, College of Dentistry, New York University, New York, New York
| | - Gary Yu
- Rory Meyers College of Nursing, New York University, New York, New York
| | - Phuong M Dong
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, California
| | - Delaney H Sheehan
- Department of Otolaryngology - Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bradley E Aouizerat
- Translational Research Center, College of Dentistry, New York University, New York, New York
| | - Carissa M Thomas
- Department of Otolaryngology - Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Chi T Viet
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, California
| | - Yi Ye
- Translational Research Center, College of Dentistry, New York University, New York, New York; Department of Molecular Pathobiology, Pain Research Center, College of Dentistry, New York University, New York, New York.
| |
Collapse
|
3
|
Liu W, Jiang H, Ke J, Liu X, Feng Y, Hou J, Long X. Changes of Trigeminal Ganglion Neurons Innervating the Temporomandibular Joint in Chronic Pain Rat Model. Int J Dent 2024; 2024:7015382. [PMID: 39309636 PMCID: PMC11415243 DOI: 10.1155/2024/7015382] [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: 07/21/2023] [Revised: 02/05/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
Background: Phenotype alterations of nociceptive neurons have been shown to be a key step in the pathogenesis of many pain-related diseases. However, it is unclear if the characteristic changes of temporomandibular joint (TMJ) primary afferent neurons are related to the pathogenesis of temporomandibular joint osteoarthritis (TMJOA) chronic pain. This study aimed to determine the morphological and neurochemical changes in trigeminal ganglion (TG) neurons innervating the TMJ in TMJOA chronic pain rats. Materials and Methods: Monosodium iodoacetate (MIA)-induced TMJOA chronic pain rat model was established (n = 6), and saline was injected in rats of the control group (n = 6). TMJ primary afferent neurons were labeled with retrograde tracing (Dil). The spatial distribution and the expression of calcitonin gene-related peptide (CGRP), isolectin B4 (IB4), and neurofilament 200 (NF200) of TMJ primary afferent neurons in TG were investigated using immunofluorescence. Intracellular calcium signaling was recorded by calcium imaging (n = 20). Results: TMJ primary afferent neurons were located only in the V3 region of the TG from both saline- and MIA-injected rats. The number of TG neurons innervating the TMJ was increased in MIA-injected rats. Elevated number and intracellular calcium concentration of small- and medium-sized instead of large-sized Dil+ TG neurons were found in MIA-injected rats. The upregulated expression of CGRP and IB4, but not NF200, in TG neurons innervating the rat TMJs was accompanied by TMJOA chronic pain. Conclusion: This study suggests that sensitization of small- to medium-sized Dil+ TG neurons and CGRP- and IB4-positive Dil+ TG neurons might contribute to the development of TMJOA chronic pain in rats. This will provide valuable information for more efficient control of TMJOA chronic pain.
Collapse
Affiliation(s)
- Wen Liu
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyGuanghua School of StomatologySun Yat-Sen University, Guangzhou, Guangdong, China
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool and Hospital of StomatologyWuhan University, Wuhan, Hubei, China
| | - Henghua Jiang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool and Hospital of StomatologyWuhan University, Wuhan, Hubei, China
| | - Jin Ke
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool and Hospital of StomatologyWuhan University, Wuhan, Hubei, China
| | - Xin Liu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool and Hospital of StomatologyWuhan University, Wuhan, Hubei, China
| | - Yaping Feng
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool and Hospital of StomatologyWuhan University, Wuhan, Hubei, China
| | - Jinsong Hou
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyGuanghua School of StomatologySun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xing Long
- State Key Laboratory of Oral and Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool and Hospital of StomatologyWuhan University, Wuhan, Hubei, China
| |
Collapse
|
4
|
Zhang FM, Wang B, Hu H, Zhang YY, Chen HH, Jiang ZJ, Zeng MX, Liu XJ. Transcriptional profiles of TGF-β superfamily members in the lumbar DRGs and the effects of activins A and C on inflammatory pain in rats. J Physiol Biochem 2023:10.1007/s13105-022-00943-z. [PMID: 36696051 DOI: 10.1007/s13105-022-00943-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/28/2022] [Indexed: 01/26/2023]
Abstract
Signaling by the transforming growth factor (TGF)-β superfamily is necessary for proper neural development and is involved in pain processing under both physiological and pathological conditions. Sensory neurons that reside in the dorsal root ganglia (DRGs) initially begin to perceive noxious signaling from their innervating peripheral target tissues and further convey pain signaling to the central nervous system. However, the transcriptional profile of the TGF-β superfamily members in DRGs during chronic inflammatory pain remains elusive. We developed a custom microarray to screen for transcriptional changes in members of the TGF-β superfamily in lumbar DRGs of rats with chronic inflammatory pain and found that the transcription of the TGF-β superfamily members tends to be downregulated. Among them, signaling of the activin/inhibin and bone morphogenetic protein/growth and differentiation factor (BMP/GDF) families dramatically decreased. In addition, peripherally pre-local administration of activins A and C worsened formalin-induced acute inflammatory pain, whereas activin C, but not activin A, improved formalin-induced persistent inflammatory pain by inhibiting the activation of astrocytes. This is the first report of the TGF-β superfamily transcriptional profiles in lumbar DRGs under chronic inflammatory pain conditions, in which transcriptional changes in cytokines or pathway components were found to contribute to, or be involved in, inflammatory pain processing. Our data will provide more targets for pain research.
Collapse
Affiliation(s)
- Feng-Ming Zhang
- School of Pharmacy, Nantong University, Jiangsu Province, 226001, Nantong, China
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, 515041, Guangdong Province, China
| | - Bing Wang
- School of Pharmacy, Nantong University, Jiangsu Province, 226001, Nantong, China
| | - Han Hu
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, No. 1 Beigou Xiangshan, Beijing, 100093, China
| | - Ying-Ying Zhang
- School of Pharmacy, Nantong University, Jiangsu Province, 226001, Nantong, China
| | - Hao-Hao Chen
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, 515041, Guangdong Province, China
| | - Zuo-Jie Jiang
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, 515041, Guangdong Province, China
| | - Mei-Xing Zeng
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, 515041, Guangdong Province, China
| | - Xing-Jun Liu
- School of Pharmacy, Nantong University, Jiangsu Province, 226001, Nantong, China.
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, 515041, Guangdong Province, China.
| |
Collapse
|
5
|
Zhang FM, Wang B, Hu H, Li QY, Chen HH, Luo LT, Jiang ZJ, Zeng MX, Liu XJ. Transcriptional Profiling of TGF-β Superfamily Members in Lumbar DRGs of Rats Following Sciatic Nerve Axotomy and Activin C Inhibits Neuropathic Pain. Endocr Metab Immune Disord Drug Targets 2023; 23:375-388. [PMID: 36201267 DOI: 10.2174/1871530322666221006114557] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 08/04/2022] [Accepted: 09/29/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Neuroinflammation and cytokines play critical roles in neuropathic pain and axon degeneration/regeneration. Cytokines of transforming growth factor-β superfamily have implications in pain and injured nerve repair processing. However, the transcriptional profiles of the transforming growth factor-β superfamily members in dorsal root ganglia under neuropathic pain and axon degeneration/regeneration conditions remain elusive. OBJECTIVE We aimed to plot the transcriptional profiles of transforming growth factor-β superfamily components in lumbar dorsal root ganglia of sciatic nerve-axotomized rats and to further verify the profiles by testing the analgesic effect of activin C, a representative cytokine, on neuropathic pain. METHODS Adult male rats were axotomized in sciatic nerves, and lumbar dorsal root ganglia were isolated for total RNA extraction or section. A custom microarray was developed and employed to plot the gene expression profiles of transforming growth factor-β superfamily components. Realtime RT-PCR was used to confirm changes in the expression of activin/inhibin family genes, and then in situ hybridization was performed to determine the cellular locations of inhibin α, activin βC, BMP-5 and GDF-9 mRNAs. The rat spared nerve injury model was performed, and a pain test was employed to determine the effect of activin C on neuropathic pain. RESULTS The expression of transforming growth factor-β superfamily cytokines and their signaling, including some receptors and signaling adaptors, were robustly upregulated. Activin βC subunit mRNAs were expressed in the small-diameter dorsal root ganglion neurons and upregulated after axotomy. Single intrathecal injection of activin C inhibited neuropathic pain in spared nerve injury model. CONCLUSION This is the first report to investigate the transcriptional profiles of members of transforming growth factor-β superfamily in axotomized dorsal root ganglia. The distinct cytokine profiles observed here might provide clues toward further study of the role of transforming growth factor-β superfamily in the pathogenesis of neuropathic pain and axon degeneration/regeneration after peripheral nerve injury.
Collapse
Affiliation(s)
- Feng-Ming Zhang
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China
| | - Bing Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Han Hu
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, No. 1 Beigou Xiangshan, Beijing, 100093, China
| | - Qing-Yi Li
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| | - Hao-Hao Chen
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| | - Li-Ting Luo
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Zuo-Jie Jiang
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| | - Mei-Xing Zeng
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| | - Xing-Jun Liu
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province, 226001, China
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, Guangdong Province, 515041, China
| |
Collapse
|
6
|
Yu X, Ton AN, Niu Z, Morales BM, Chen J, Braz J, Lai MH, Barruet E, Liu H, Cheung K, Ali S, Chan T, Bigay K, Ho J, Nikolli I, Hansberry S, Wentworth K, Kriegstein A, Basbaum A, Hsiao EC. ACVR1-activating mutation causes neuropathic pain and sensory neuron hyperexcitability in humans. Pain 2023; 164:43-58. [PMID: 35442931 PMCID: PMC9582048 DOI: 10.1097/j.pain.0000000000002656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 03/01/2022] [Accepted: 04/08/2022] [Indexed: 01/09/2023]
Abstract
ABSTRACT Altered bone morphogenetic protein (BMP) signaling is associated with many musculoskeletal diseases. However, it remains unknown whether BMP dysfunction has direct contribution to debilitating pain reported in many of these disorders. Here, we identified a novel neuropathic pain phenotype in patients with fibrodysplasia ossificans progressiva (FOP), a rare autosomal-dominant musculoskeletal disorder characterized by progressive heterotopic ossification. Ninety-seven percent of these patients carry an R206H gain-of-function point mutation in the BMP type I receptor ACVR1 (ACVR1 R206H ), which causes neofunction to Activin A and constitutively activates signaling through phosphorylated SMAD1/5/8. Although patients with FOP can harbor pathological lesions in the peripheral and central nervous system, their etiology and clinical impact are unclear. Quantitative sensory testing of patients with FOP revealed significant heat and mechanical pain hypersensitivity. Although there was no major effect of ACVR1 R206H on differentiation and maturation of nociceptive sensory neurons (iSNs) derived from FOP induced pluripotent stem cells, both intracellular and extracellular electrophysiology analyses of the ACVR1 R206H iSNs displayed ACVR1-dependent hyperexcitability, a hallmark of neuropathic pain. Consistent with this phenotype, we recorded enhanced responses of ACVR1 R206H iSNs to TRPV1 and TRPA1 agonists. Thus, activated ACVR1 signaling can modulate pain processing in humans and may represent a potential target for pain management in FOP and related BMP pathway diseases.
Collapse
Affiliation(s)
- Xiaobing Yu
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States
| | - Amy N. Ton
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Zejun Niu
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States
- Department of Anesthesiology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Blanca M. Morales
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Jiadong Chen
- Department of Neurology, University of California, San Francisco, CA, United States. Dr. Chen is now with the Department of Neurology of Second Affiliated Hospital, Centre for Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Joao Braz
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| | - Michael H. Lai
- J. David Gladstone Institutes, San Francisco, CA, United States
| | - Emilie Barruet
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Hongju Liu
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States
- Department of Anesthesiology, Peking Union Medical College Hospital, Beijing, China
| | - Kin Cheung
- BioSAS Consulting, Inc, Wellesley, MA, United States
| | - Syed Ali
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States
| | - Tea Chan
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Katherine Bigay
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Jennifer Ho
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Ina Nikolli
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Steven Hansberry
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
- California Institute of Regenerative Medicine Bridges to Stem Cell Research Program, San Francisco State University, San Francisco, CA, United States
| | - Kelly Wentworth
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Arnold Kriegstein
- Department of Neurology, University of California, San Francisco, CA, United States. Dr. Chen is now with the Department of Neurology of Second Affiliated Hospital, Centre for Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Allan Basbaum
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| | - Edward C. Hsiao
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| |
Collapse
|
7
|
Dubeykovskaya ZA, Tu NH, Garcia PDR, Schmidt BL, Albertson DG. Oral Cancer Cells Release Vesicles that Cause Pain. Adv Biol (Weinh) 2022; 6:e2200073. [PMID: 35802912 PMCID: PMC9474716 DOI: 10.1002/adbi.202200073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/02/2022] [Indexed: 01/28/2023]
Abstract
Oral cancer pain is attributed to the release from cancers of mediators that sensitize and activate sensory neurons. Intraplantar injection of conditioned media (CM) from human tongue cancer cell line HSC-3 or OSC-20 evokes nociceptive behavior. By contrast, CM from noncancer cell lines, DOK, and HaCaT are non-nociceptive. Pain mediators are carried by extracellular vesicles (EVs) released from cancer cells. Depletion of EVs from cancer cell line CM reverses mechanical allodynia and thermal hyperalgesia. CM from non-nociceptive cell lines become nociceptive when reconstituted with HSC-3 EVs. Two miRNAs (hsa-miR-21-5p and hsa-miR-221-3p) are identified that are present in increased abundance in EVs from HSC-3 and OSC-20 CM compared to HaCaT CM. The miRNA target genes suggest potential involvement in oral cancer pain of the toll like receptor 7 (TLR7) and 8 (TLR8) pathways, as well as signaling through interleukin 6 cytokine family signal transducer receptor (gp130, encoded by IL6ST) and colony stimulating factor receptor (G-CSFR, encoded by CSF3R), Janus kinase and signal transducer and activator of transcription 3 (JAK/STAT3). These studies confirm the recent discovery of the role of cancer EVs in pain and add to the repertoire of algesic and analgesic cancer pain mediators and pathways that contribute to oral cancer pain.
Collapse
Affiliation(s)
- Zinaida A Dubeykovskaya
- Bluestone Center for Clinical Research and Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Nguyen Huu Tu
- Bluestone Center for Clinical Research and Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Paulina D Ramírez Garcia
- Bluestone Center for Clinical Research and Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Brian L Schmidt
- Bluestone Center for Clinical Research and Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Donna G Albertson
- Bluestone Center for Clinical Research and Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| |
Collapse
|
8
|
Lin W, Zhang WW, Lyu N, Cao H, Xu WD, Zhang YQ. Growth Differentiation Factor-15 Produces Analgesia by Inhibiting Tetrodotoxin-Resistant Nav1.8 Sodium Channel Activity in Rat Primary Sensory Neurons. Neurosci Bull 2021; 37:1289-1302. [PMID: 34076854 PMCID: PMC8423960 DOI: 10.1007/s12264-021-00709-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/10/2021] [Indexed: 01/01/2023] Open
Abstract
Growth differentiation factor 15 (GDF-15) is a member of the transforming growth factor-β superfamily. It is widely distributed in the central and peripheral nervous systems. Whether and how GDF-15 modulates nociceptive signaling remains unclear. Behaviorally, we found that peripheral GDF-15 significantly elevated nociceptive response thresholds to mechanical and thermal stimuli in naïve and arthritic rats. Electrophysiologically, we demonstrated that GDF-15 decreased the excitability of small-diameter dorsal root ganglia (DRG) neurons. Furthermore, GDF-15 concentration-dependently suppressed tetrodotoxin-resistant sodium channel Nav1.8 currents, and shifted the steady-state inactivation curves of Nav1.8 in a hyperpolarizing direction. GDF-15 also reduced window currents and slowed down the recovery rate of Nav1.8 channels, suggesting that GDF-15 accelerated inactivation and slowed recovery of the channel. Immunohistochemistry results showed that activin receptor-like kinase-2 (ALK2) was widely expressed in DRG medium- and small-diameter neurons, and some of them were Nav1.8-positive. Blockade of ALK2 prevented the GDF-15-induced inhibition of Nav1.8 currents and nociceptive behaviors. Inhibition of PKA and ERK, but not PKC, blocked the inhibitory effect of GDF-15 on Nav1.8 currents. These results suggest a functional link between GDF-15 and Nav1.8 in DRG neurons via ALK2 receptors and PKA associated with MEK/ERK, which mediate the peripheral analgesia of GDF-15.
Collapse
Affiliation(s)
- Wei Lin
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Wen-Wen Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ning Lyu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Hong Cao
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Wen-Dong Xu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China. .,Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Yu-Qiu Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China. .,Department of Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
9
|
Cardiopulmonary and Neurologic Dysfunctions in Fibrodysplasia Ossificans Progressiva. Biomedicines 2021; 9:biomedicines9020155. [PMID: 33562570 PMCID: PMC7915901 DOI: 10.3390/biomedicines9020155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/20/2021] [Accepted: 01/28/2021] [Indexed: 12/28/2022] Open
Abstract
Fibrodysplasia Ossificans Progressiva (FOP) is an ultra-rare but debilitating disorder characterized by spontaneous, progressive, and irreversible heterotopic ossifications (HO) at extraskeletal sites. FOP is caused by gain-of-function mutations in the Activin receptor Ia/Activin-like kinase 2 gene (Acvr1/Alk2), with increased receptor sensitivity to bone morphogenetic proteins (BMPs) and a neoceptor response to Activin A. There is extensive literature on the skeletal phenotypes in FOP, but a much more limited understanding of non-skeletal manifestations of this disease. Emerging evidence reveals important cardiopulmonary and neurologic dysfunctions in FOP including thoracic insufficiency syndrome, pulmonary hypertension, conduction abnormalities, neuropathic pain, and demyelination of the central nervous system (CNS). Here, we review the recent research and discuss unanswered questions regarding the cardiopulmonary and neurologic phenotypes in FOP.
Collapse
|
10
|
Bhattacharya A, Janal MN, Veeramachaneni R, Dolgalev I, Dubeykovskaya Z, Tu NH, Kim H, Zhang S, Wu AK, Hagiwara M, Kerr AR, DeLacure MD, Schmidt BL, Albertson DG. Oncogenes overexpressed in metastatic oral cancers from patients with pain: potential pain mediators released in exosomes. Sci Rep 2020; 10:14724. [PMID: 32895418 PMCID: PMC7477576 DOI: 10.1038/s41598-020-71298-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 08/10/2020] [Indexed: 12/11/2022] Open
Abstract
Oral cancer patients experience pain at the site of the primary cancer. Patients with metastatic oral cancers report greater pain. Lack of pain identifies patients at low risk of metastasis with sensitivity = 0.94 and negative predictive value = 0.89. In the same cohort, sensitivity and negative predictive value of depth of invasion, currently the best predictor, were 0.95 and 0.92, respectively. Cancer pain is attributed to cancer-derived mediators that sensitize neurons and is associated with increased neuronal density. We hypothesized that pain mediators would be overexpressed in metastatic cancers from patients reporting high pain. We identified 40 genes overexpressed in metastatic cancers from patients reporting high pain (n = 5) compared to N0 cancers (n = 10) and normal tissue (n = 5). The genes are enriched for functions in extracellular matrix organization and angiogenesis. They have oncogenic and neuronal functions and are reported in exosomes. Hierarchical clustering according to expression of neurotrophic and axon guidance genes also separated cancers according to pain and nodal status. Depletion of exosomes from cancer cell line supernatant reduced nociceptive behavior in a paw withdrawal assay, supporting a role for exosomes in cancer pain. The identified genes and exosomes are potential therapeutic targets for stopping cancer and attenuating pain.
Collapse
Affiliation(s)
- Aditi Bhattacharya
- Bluestone Center for Clinical Research, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY, 10010, USA.,Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Malvin N Janal
- Department of Epidemiology and Health Promotion, New York University College of Dentistry, New York, NY, 10010, USA
| | - Ratna Veeramachaneni
- Bluestone Center for Clinical Research, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY, 10010, USA.,Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Igor Dolgalev
- Applied Bioinformatics Laboratories, New York University Langone Medical Center, New York, NY, 10016, USA
| | - Zinaida Dubeykovskaya
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Nguyen Huu Tu
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Hyesung Kim
- New York University College of Dentistry, New York, NY, 10010, USA
| | - Susanna Zhang
- New York University College of Dentistry, New York, NY, 10010, USA
| | - Angie K Wu
- Bluestone Center for Clinical Research, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY, 10010, USA
| | - Mari Hagiwara
- Department of Radiology, New York University School of Medicine, New York, NY, 10016, USA
| | - A Ross Kerr
- Department of Oral and Maxillofacial Pathology, Radiology and Medicine, New York University, New York, NY, 10010, USA
| | - Mark D DeLacure
- Division of Head and Neck Surgery and Oncology, New York University School of Medicine, New York, NY, 10016, USA
| | - Brian L Schmidt
- Bluestone Center for Clinical Research, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY, 10010, USA.,Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Donna G Albertson
- Bluestone Center for Clinical Research, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY, 10010, USA. .,Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA.
| |
Collapse
|
11
|
Liu Z, Murphy SF, Huang J, Zhao L, Hall CC, Schaeffer AJ, Schaeffer EM, Thumbikat P. A novel immunocompetent model of metastatic prostate cancer-induced bone pain. Prostate 2020; 80:782-794. [PMID: 32407603 PMCID: PMC7375026 DOI: 10.1002/pros.23993] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 03/18/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Over 70% to 85% of men with advanced prostate cancer (PCa) develop bone metastases characterized by severe bone pain and increased likelihood of bone fracture. These clinical features result in decreased quality of life and act as a predictor of higher mortality. Mechanistically, the skeletal pathologies such as osteolytic lesions and abnormal osteoblastic activity drive these symptoms. The role of immune cells in bone cancer pain remains understudied, here we sought to recapitulate this symptomology in a murine model. METHODS The prostate cancer bone metastasis-induced pain model (CIBP) was established by transplanting a mouse prostate cancer cell line into the femur of immunocompetent mice. Pain development, gait dynamics, and the changes in emotional activities like depression and anxiety were evaluated. Animal tissues including femurs, dorsal root ganglion (DRG), and spinal cord were collected at killing and microcomputed tomography (μCT), histology/immunohistochemistry, and quantitative immunofluorescent analysis were performed. RESULTS Mice receiving prostate cancer cells showed a significantly lower threshold for paw withdrawal responses induced by mechanical stimulation compared with their control counterparts. Zero maze and DigiGait analyses indicated reduced and aberrant movement associated emotional activity compared with sham control at 8-weeks postinjection. The μCT analysis showed osteolytic and osteoblastic changes and a 50% reduction of the trabecular volumes within the prostate cancer group. Neurologically we demonstrated, increased calcitonin gene-related peptide (CGRP) and neuronal p75NTR immune-reactivities in both the projected terminals of the superficial dorsal horn and partial afferent neurons in DRG at L2 to L4 level in tumor-bearing mice. Furthermore, our data show elevated nerve growth factor (NGF) and TrkA immunoreactivities in the same segment of the superficial dorsal horn that were, however, not colocalized with CGRP and p75NTR . CONCLUSIONS This study describes a novel immunocompetent model of CIBP and demonstrates the contribution of NGF and p75NTR to chronic pain in bone metastasis.
Collapse
Affiliation(s)
- Zhiqiang Liu
- Dept. of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Stephen F. Murphy
- Dept. of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Jian Huang
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, USA
| | - Lan Zhao
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, USA
| | - Christel C. Hall
- Dept. of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Anthony J. Schaeffer
- Dept. of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Edward M. Schaeffer
- Dept. of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Praveen Thumbikat
- Dept. of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
12
|
Tang R, Harasymowicz NS, Wu CL, Collins KH, Choi YR, Oswald SJ, Guilak F. Gene therapy for follistatin mitigates systemic metabolic inflammation and post-traumatic arthritis in high-fat diet-induced obesity. SCIENCE ADVANCES 2020; 6:eaaz7492. [PMID: 32426485 PMCID: PMC7209997 DOI: 10.1126/sciadv.aaz7492] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 02/26/2020] [Indexed: 05/11/2023]
Abstract
Obesity-associated inflammation and loss of muscle function play critical roles in the development of osteoarthritis (OA); thus, therapies that target muscle tissue may provide novel approaches to restoring metabolic and biomechanical dysfunction associated with obesity. Follistatin (FST), a protein that binds myostatin and activin, may have the potential to enhance muscle formation while inhibiting inflammation. Here, we hypothesized that adeno-associated virus 9 (AAV9) delivery of FST enhances muscle formation and mitigates metabolic inflammation and knee OA caused by a high-fat diet in mice. AAV-mediated FST delivery exhibited decreased obesity-induced inflammatory adipokines and cytokines systemically and in the joint synovial fluid. Regardless of diet, mice receiving FST gene therapy were protected from post-traumatic OA and bone remodeling induced by joint injury. Together, these findings suggest that FST gene therapy may provide a multifactorial therapeutic approach for injury-induced OA and metabolic inflammation in obesity.
Collapse
Affiliation(s)
- Ruhang Tang
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Natalia S. Harasymowicz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Yun-Rak Choi
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Sara J. Oswald
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| |
Collapse
|
13
|
Qiu CY, Liu TT, Wei S, Zhou YM, Wu L, Jin Y, Hu WP. TGF-β1 enhances the activity of acid-sensing ion channel in rat primary sensory neurons. J Neurosci Res 2019; 97:1298-1305. [PMID: 31240740 DOI: 10.1002/jnr.24481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/20/2022]
Abstract
Transforming growth factor-β1 (TGF-β1) is an important member of multifunctional growth factor superfamily. It has been implicated in pain signaling, but little is known about the underlying mechanisms. Herein, we report that TGF-β1 can exert a sustained enhancing effect on the functional activity of acid-sensing ion channels (ASICs) in rat dorsal root ganglia (DRG) neurons. Pre-application of TGF-β1 increased the amplitude of proton-gated currents in a dose-dependent manner. Enhancement of ASIC currents lasted for more than 30 min although TGF-β1 was treated once only. This sustained enhancement by TGF-β1 could be blocked by extracellular treatment of selective TGF-β receptor I antagonist SD-208, and abolished by blockade of intracellular several non-Smad-signaling pathways. TGF-β1 also sustainedly enhanced proton-evoked spikes in rat DRG neurons. Moreover, peripheral pre-treatment with TGF-β1 dose-dependently exacerbated nociceptive behaviors evoked by intraplantar injection of acetic acid through TGF-β receptor I in rats. These results suggested that TGF-β1 potentiated ASIC-mediated electrophysiological activity and nociceptive behaviors, which revealed a novel mechanism underlying TGF-β1 implicated in peripheral pain signaling by sensitizing ASICs.
Collapse
Affiliation(s)
- Chun-Yu Qiu
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, P R China.,Department of Pharmacology, Hubei University of Science and Technology, Xianning, P R China
| | - Ting-Ting Liu
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, P R China
| | - Shuang Wei
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, P R China
| | - Yi-Mei Zhou
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, P R China
| | - Lei Wu
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, P R China
| | - Ying Jin
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, P R China
| | - Wang-Ping Hu
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, P R China
| |
Collapse
|
14
|
Yu H, Shin SM, Wang F, Xu H, Xiang H, Cai Y, Itson-Zoske B, Hogan QH. Transmembrane protein 100 is expressed in neurons and glia of dorsal root ganglia and is reduced after painful nerve injury. Pain Rep 2018; 4:e703. [PMID: 30801043 PMCID: PMC6370145 DOI: 10.1097/pr9.0000000000000703] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 10/08/2018] [Accepted: 10/30/2018] [Indexed: 12/16/2022] Open
Abstract
Introduction Tmem100 modulates interactions between TRPA1 and TRPV1. The cell specificity of Tmem100 expression in dorsal root ganglia (DRGs) is not well defined, nor is the effect of peripheral nerve injury on Tmem100 expression. Objective This study was designed to determine the cell specificity of Tmem100 expression in DRG and its subcellular localization, and to examine how Tmem100 expression may be altered in painful conditions. Methods Dorsal root ganglion Tmem100 expression was determined by immunohistochemistry, immunoblot, and quantitative real-time PCR, and compared between various experimental rat pain models and controls. Results Tmem100 is expressed in both neurons and perineuronal glial cells in the rat DRG. The plasma membrane and intracellular localization of Tmem100 are identified in 83% ± 6% of IB4-positive and 48% ± 6% of calcitonin gene-related peptide-positive neurons, as well as in medium- and large-sized neurons, with its immunopositivity colocalized to TRPV1 (94% ± 5%) and TRPA1 (96% ± 3%). Tmem100 is also detected in the perineuronal satellite glial cells and in some microglia. Tmem100 protein is significantly increased in the lumbar DRGs in the complete Freund adjuvant inflammatory pain. By contrast, peripheral nerve injury by spinal nerve ligation diminishes Tmem100 expression in the injured DRG, with immunoblot and immunohistochemistry experiments showing reduced Tmem100 protein levels in both neurons and satellite glial cells of DRGs proximal to injury, whereas Tmem100 is unchanged in adjacent DRGs. The spared nerve injury model also reduces Tmem100 protein in the injured DRGs. Conclusion Our data demonstrate a pain pathology-dependent alteration of DRG Tmem100 protein expression, upregulated during CFA inflammatory pain but downregulated during neuropathic pain.
Collapse
Affiliation(s)
- Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
| | - Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Fei Wang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, PR of China
| | - Hao Xu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, PR of China
| | - Hongfei Xiang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, PR of China
| | - Yongsong Cai
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR of China
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
| |
Collapse
|
15
|
Presynaptic inhibition of nociceptive neurotransmission by somatosensory neuron-secreted suppressors. SCIENCE CHINA-LIFE SCIENCES 2017. [PMID: 28624955 DOI: 10.1007/s11427-017-9061-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Noxious stimuli cause pain by activating cutaneous nociceptors. The Aδ- and C-fibers of dorsal root ganglion (DRG) neurons convey the nociceptive signals to the laminae I-II of spinal cord. In the dorsal horn of spinal cord, the excitatory afferent synaptic transmission is regulated by the inhibitory neurotransmitter γ-aminobutyric acid and modulators such as opioid peptides released from the spinal interneurons, and by serotonin, norepinepherine and dopamine from the descending inhibitory system. In contrast to the accumulated evidence for these central inhibitors and their neural circuits in the dorsal spinal cord, the knowledge about the endogenous suppressive mechanisms in nociceptive DRG neurons remains very limited. In this review, we summarize our recent findings of the presynaptic suppressive mechanisms in nociceptive neurons, the BNP/NPR-A/PKG/BKCa channel pathway, the FSTL1/α1Na+-K+ ATPase pathway and the activin C/ERK pathway. These endogenous suppressive systems in the mechanoheat nociceptors may also contribute differentially to the mechanisms of nerve injury-induced neuropathic pain or inflammation-induced pain.
Collapse
|
16
|
Tanaka K, Kuwahara-Otani S, Maeda S, Minato Y, Yagi H. Possible Role of the Myelinated Neural Network in the Parietal Peritoneum in Rats as a Mechanoreceptor. Anat Rec (Hoboken) 2017; 300:1662-1669. [PMID: 28524374 DOI: 10.1002/ar.23613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/19/2016] [Accepted: 12/27/2016] [Indexed: 12/24/2022]
Abstract
A network of myelinated nerve fibers in the peritoneum covers the abdominal wall. We studied the topographic distribution of this network, explored the fibers' destination in the central nervous system, and examined the markers in these fibers in order to identify the nature of the sensation conveyed by the network of nerve fibers in rats. We used Sihler's method, which stains myelinated fibers in whole mount materials, and observed a dense nerve network and endings toward the peritoneal cavity in the peritoneum that covers the abdomen's lateral bulge. We studied the axonal transport of cholera toxin subunit B to investigate the central projections of this network in order to identify its function. After applying the tracer in the peritoneum, we observed many labeled terminals in the medial part of laminae 3-5 of the spinal cord. A small number of labeled terminals was observed in the dorsal nucleus of Clarke and gracile nucleus. Labeled somata were observed in the dorsal root ganglia (DRG). Most (96%) were larger than 35 μm. We performed immunohistochemistry of the abdominal wall, using antiserum against the 200-kD neurofilament (a marker for mechanosensory neurons). We observed many positive nerve fibers in the peritoneum. Because cell bodies in the DRG were large, their nerve terminals ended in the base of the dorsal horn, which is known to transmit proprioceptive information, and the network possesses the marker for mechanosensitive fibers; therefore, it appears that the myelinated nerve network conveys information about distension and/or contraction of the abdominal wall. Anat Rec, 300:1662-1669, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Koichi Tanaka
- Department of Anatomy, Division of Cell Biology, Hyogo College of Medicine, Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Sachi Kuwahara-Otani
- Department of Anatomy, Division of Cell Biology, Hyogo College of Medicine, Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Seishi Maeda
- Department of Anatomy, Division of Cell Biology, Hyogo College of Medicine, Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Yusuke Minato
- Department of Anatomy, Division of Cell Biology, Hyogo College of Medicine, Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Hideshi Yagi
- Department of Anatomy, Division of Cell Biology, Hyogo College of Medicine, Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| |
Collapse
|
17
|
A research design for the quantification of the neuropeptides substance p and calcitonin gene-related Peptide in rat skin using Western blot analysis. Adv Skin Wound Care 2016; 28:259-65. [PMID: 25988735 DOI: 10.1097/01.asw.0000465373.42350.c1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To describe and standardize a protocol that overcomes the technical limitations of Western blot (WB) analysis in the quantification of the neuropeptides substance P (SP) and calcitonin gene-related peptide (CGRP) following nociceptive stimuli in rat skin. DESIGN Male Wistar rats (Rattus norvegicus albinus) weighing 250 to 350 g were used in this study. Elements of WB analysis were adapted by using specific manipulation of samples, repeated cycles of freezing and thawing, more thorough maceration, and a more potent homogenizer; increasing lytic reagents; promoting greater inhibition of protease activity; and using polyvinylidene fluoride membranes as transfer means for skin-specific protein. Other changes were also made to adapt the WB analysis to a rat model. SETTING University research center. MAIN OUTCOME MEASURE Western blot analysis adapted to a rat model. RESULTS This research design has proven effective in collecting and preparing skin samples to quantify SP and CGRP using WB analysis in rat skin. CONCLUSION This study described a research design that uses WB analysis as a reproducible, technically accessible, and cost-effective method for the quantification of SP and CGRP in rat skin that overcomes technical biases.
Collapse
|
18
|
Mitchell K, Shah JP, Dalgard CL, Tsytsikova LV, Tipton AC, Dmitriev AE, Symes AJ. Bone morphogenetic protein-2-mediated pain and inflammation in a rat model of posterolateral arthrodesis. BMC Neurosci 2016; 17:80. [PMID: 27905881 PMCID: PMC5134101 DOI: 10.1186/s12868-016-0314-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 11/24/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Bone morphogenetic protein-2 (BMP-2) is a pleiotropic, secreted molecule with diverse effects. The potent ability of BMP-2 to stimulate bone growth prompted its widespread clinical use for arthrodesis (spine fusion). However, elevated post-operative pain in patients treated with BMP-2 has been increasingly reported. Determining whether BMP-2 induces pain directly or whether it induces neuroinflammation, which could lower the threshold for pain, is important for developing therapeutic interventions. We therefore modeled the clinical use of BMP-2 for posterior lumbar fusion by implanting absorbable collagen sponges soaked with either recombinant human BMP-2 (rhBMP-2) or vehicle above the L4-L5 transverse processes of rat spine. RESULTS Using microarray analysis we found that implantation of rhBMP-2-soaked absorbable collagen sponges resulted in altered expression of numerous pro-inflammatory genes in the adjacent dorsal root ganglia (DRG) showing that implantation of rhBMP-2/absorbable collagen sponges triggers potent neuroinflammatory responses in the DRG-2. Interestingly, direct BMP-2 treatment of DRG explants resulted in changes in gene expression that were not specifically pro-inflammatory. Rats implanted with rhBMP-2 in absorbable collagen sponges also exhibited a transient change in thermal and mechanical sensitivity indicating that rhBMP-2 applied to the lumbar spine could increase pain sensitivity. Immunohistochemical analysis indicated macrophage infiltration in the DRG and spinal nerve in rats implanted with rhBMP-2/absorbable collagen sponges or absorbable collagen sponges alone, but not in rats that underwent surgery without implantation of the absorbable collagen sponges suggesting that the sponges contributed to the biological response. Indeed, analysis of DRGs taken from rats implanted with absorbable collagen sponges without rhBMP-2 showed a significant change in gene expression distinct from DRGs from rats undergoing surgery only. CONCLUSIONS Our data indicate that implantation of rhBMP-2/absorbable collagen sponges on the lumbar spine triggers potent neuroinflammatory responses in the DRG. Importantly, however, these BMP-2 effects may be partially mediated through a response to the absorbable collagen sponges.
Collapse
Affiliation(s)
- Kendall Mitchell
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Jill P Shah
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, MD, USA
| | - Lyubov V Tsytsikova
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Ashley C Tipton
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Anton E Dmitriev
- Department of Surgery, Uniformed Services University, Bethesda, MD, USA.,Department of Orthopaedic Surgery, Walter Reed National Military Medical Center, Bethesda, MD, 20814, USA.,Division of Applied Mechanics, CDRH/OSEL, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Aviva J Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
19
|
Hashmi F, Liu M, Shen S, Qiao LY. EXPRESS: Phospholipase C gamma mediates endogenous brain-derived neurotrophic factor - regulated calcitonin gene-related peptide expression in colitis - induced visceral pain. Mol Pain 2016; 12:12/0/1744806916657088. [PMID: 27306412 PMCID: PMC4955977 DOI: 10.1177/1744806916657088] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Visceral hypersensitivity is a complex pathophysiological paradigm with unclear mechanisms. Primary afferent neuronal plasticity marked by alterations in neuroactive compounds such as calcitonin gene-related peptide is suggested to underlie the heightened sensory responses. Signal transduction that leads to calcitonin gene-related peptide expression thereby sensory neuroplasticity during colitis remains to be elucidated. RESULTS In a rat model with colitis induced by 2,4,6-trinitrobenzene sulfonic acid, we found that endogenously elevated brain-derived neurotrophic factor elicited an up-regulation of calcitonin gene-related peptide in the lumbar L1 dorsal root ganglia. At seven days of colitis, neutralization of brain-derived neurotrophic factor with a specific brain-derived neurotrophic factor antibody reversed calcitonin gene-related peptide up-regulation in the dorsal root ganglia. Colitis-induced calcitonin gene-related peptide transcription was also inhibited by brain-derived neurotrophic factor antibody treatment. Signal transduction studies with dorsal root ganglia explants showed that brain-derived neurotrophic factor-induced calcitonin generelated peptide expression was mediated by the phospholipase C gamma, but not the phosphatidylinositol 3-kinase/Akt or the mitogen-activated protein kinase/extracellular signal-regulated protein kinase pathway. Application of PLC inhibitor U73122 in vivo confirmed that colitis-induced and brain-derived neurotrophic factor-mediated calcitonin gene-related peptide up-regulation in the dorsal root ganglia was regulated by the phospholipase C gamma pathway. In contrast, suppression of the phosphatidylinositol 3-kinase activity in vivo had no effect on colitis-induced calcitonin gene-related peptide expression. During colitis, calcitonin gene-related peptide also co-expressed with phospholipase C gamma but not with p-Akt. Calcitonin gene-related peptide up-regulation during colitis correlated to the activation of cAMP-responsive element binding protein in the same neurons. Consistently, colitis-induced cAMP-responsive element binding protein activation in the dorsal root ganglia was attenuated by brain-derived neurotrophic factor antibody treatment. CONCLUSION These results suggest that colitis-induced and brain-derived neurotrophic factor-mediated calcitonin generelated peptide expression in sensory activation is regulated by a unique pathway involving brain-derived neurotrophic factorphospholipase C gamma-cAMP-responsive element binding protein axis.
Collapse
Affiliation(s)
- Fiza Hashmi
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Miao Liu
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Shanwei Shen
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Li-Ya Qiao
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Li-Ya Qiao, Department of Physiology and Biophysics, PO Box 980551, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298–0551, USA.
| |
Collapse
|
20
|
Shaffer AD, Feng B, La JH, Joyce SC, Gebhart GF. A novel role for follistatin in hypersensitivity following cystitis. Neurourol Urodyn 2015; 36:286-292. [PMID: 26713850 DOI: 10.1002/nau.22941] [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: 08/19/2015] [Accepted: 11/23/2015] [Indexed: 11/09/2022]
Abstract
AIMS Previous studies have shown that the activin-binding protein follistatin reduces inflammation in several mouse models of colitis. To determine whether follistatin also has a beneficial effect following bladder inflammation, we induced cystitis in mice using cyclophosphamide (CYP) and examined the relationship between bladder hypersensitivity and bladder follistatin expression. METHODS Adult female C57BL/6 mice were treated with CYP (100 mg/kg) or vehicle (saline) three times over 5 days. Bladder hypersensitivity was assessed by recording the visceromotor response (VMR) to urinary bladder distension and in vitro single-fiber bladder afferent recording. Follistatin gene expression was measured using qRT-PCR. Immunohistochemistry was employed for further characterization. RESULTS Bladder hypersensitivity was established by day 6 and persisted to day 14 in CYP-treated mice. On day 14, hypersensitivity was accompanied by increases in follistatin gene expression in the bladder. Follistatin-like immunoreactivity colocalized with laminin, and the percentage of structures in the lamina propria that were follistatin-positive was increased in CYP-treated mice. Exogenous follistatin increased VMR and afferent responses to bladder distension in CYP- but not vehicle-treated mice. CONCLUSIONS Chronic bladder pain following CYP treatment is associated with increased follistatin expression in the bladder. These results suggest a novel, pro-nociceptive role for follistatin in cystitis, in contrast with its proposed therapeutic role in colitis. This protein has exciting potential as a biomarker and therapeutic target for bladder hypersensitivity. Neurourol. Urodynam. 36:286-292, 2017. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amber D Shaffer
- Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bin Feng
- Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jun-Ho La
- Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sonali C Joyce
- Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - G F Gebhart
- Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
21
|
Tillmaand EG, Yang N, Kindt CAC, Romanova EV, Rubakhin SS, Sweedler JV. Peptidomics and Secretomics of the Mammalian Peripheral Sensory-Motor System. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2015; 26:2051-2061. [PMID: 26392278 PMCID: PMC4655166 DOI: 10.1007/s13361-015-1256-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/30/2015] [Accepted: 08/10/2015] [Indexed: 06/01/2023]
Abstract
The dorsal root ganglion (DRG) and its anatomically and functionally associated spinal nerve and ventral and dorsal roots are important components of the peripheral sensory-motor system in mammals. The cells within these structures use a number of peptides as intercellular signaling molecules. We performed a variety of mass spectrometry (MS)-based characterizations of peptides contained within and secreted from these structures, and from isolated and cultured DRG cells. Liquid chromatography-Fourier transform MS was utilized in DRG and nerve peptidome analysis. In total, 2724 peptides from 296 proteins were identified in tissue extracts. Neuropeptides are among those detected, including calcitonin gene-related peptide I, little SAAS, and known hemoglobin-derived peptides. Solid phase extraction combined with direct matrix-assisted laser desorption/ionization time-of-flight MS was employed to investigate the secretome of these structures. A number of peptides were detected in the releasate from semi-intact preparations of DRGs and associated nerves, including neurofilament- and myelin basic protein-related peptides. A smaller set of analytes was observed in releasates from cultured DRG neurons. The peptide signals observed in the releasates have been mass-matched to those characterized and identified in homogenates of entire DRGs and associated nerves. This data aids our understanding of the chemical composition of the mammalian peripheral sensory-motor system, which is involved in key physiological functions such as nociception, thermoreception, itch sensation, and proprioception.
Collapse
Affiliation(s)
- Emily G Tillmaand
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Ning Yang
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Callie A C Kindt
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Elena V Romanova
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Stanislav S Rubakhin
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jonathan V Sweedler
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
22
|
Schüttenhelm BN, Duraku LS, Dijkstra JF, Walbeehm ET, Holstege JC. Differential Changes in the Peptidergic and the Non-Peptidergic Skin Innervation in Rat Models for Inflammation, Dry Skin Itch, and Dermatitis. J Invest Dermatol 2015; 135:2049-2057. [PMID: 25848979 DOI: 10.1038/jid.2015.137] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 11/25/2014] [Accepted: 12/11/2014] [Indexed: 01/07/2023]
Abstract
Skin innervation is a dynamic process that may lead to changes in nerve fiber density during pathological conditions. We have investigated changes in epidermal nerve fiber density in three different rat models that selectively produce chronic itch (the dry skin model), or itch and inflammation (the dermatitis model), or chronic inflammation without itch (the CFA model). In the epidermis, we identified peptidergic fibers-that is, immunoreactive (IR) for calcitonin gene-related peptide or substance P—and non-peptidergic fibers—that is, IR for P2X3. The overall density of nerve fibers was determined using IR for the protein gene product 9.5. In all three models, the density of epidermal peptidergic nerve fibers increased up to five times when compared with a sham-treated control group. In contrast, the density of epidermal non-peptidergic fibers was not increased, except for a small but significant increase in the dry skin model. Chronic inflammation showed an increased density of peptidergic fibers without itch, indicating that increased nerve fiber density is not invariably associated with itch. The finding that different types of skin pathology induced differential changes in nerve fiber density may be used as a diagnostic tool in humans, through skin biopsies, to identify different types of pathology and to monitor the effect of therapies.
Collapse
Affiliation(s)
- Barthold N Schüttenhelm
- Department of Neuroscience, Erasmus Medical Centre, Rotterdam, The Netherlands; Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Liron S Duraku
- Department of Neuroscience, Erasmus Medical Centre, Rotterdam, The Netherlands; Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Jouke F Dijkstra
- Department of Neuroscience, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Erik T Walbeehm
- Department of Plastic Surgery, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Jan C Holstege
- Department of Neuroscience, Erasmus Medical Centre, Rotterdam, The Netherlands.
| |
Collapse
|
23
|
Calcitonin gene-related peptide is involved in inflammatory pain but not in postoperative pain. Anesthesiology 2014; 121:1068-79. [PMID: 24992521 DOI: 10.1097/aln.0000000000000364] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The aim of this study was to clarify the roles of calcitonin gene-related peptide (CGRP) in postoperative pain and inflammatory pain. METHODS αCGRP knockout mice that the authors have developed and wild-type mice were used. Pain behaviors were assessed after incision and complete Freund's adjuvant (CFA) injection. Changes in CGRP and c-Fos expression in the dorsal horn were also examined. RESULTS Guarding pain scores in αCGRP knockout mice were lower than those in wild-type mice at 24 h (3.8 ± 1.6 vs. 6.8 ± 1.5, P = 0.044) and 48 h (1.8 ± 1.7 vs. 6.0 ± 1.5, P = 0.001) after CFA injection (n = 8 to 9). Withdrawal latencies to heat stimulation in αCGRP knockout mice were higher than those in wild-type mice at 24 to 72 h after CFA injection (4.9 ± 1.0 vs. 3.4 ± 0.8 at 24 h, P = 0.04; 5.1 ± 0.3 vs. 3.2 ± 0.9 at 48 h, P = 0.047; and 5.4 ± 1.6 vs. 3.5 ± 0.5 s at 72 h, P = 0.045) (n = 11 to 13), but withdrawal thresholds to mechanical stimulation were comparable. CGRP expression was increased at 24 h after CFA injection in wild-type mice, and the c-Fos-positive profile was increased at 4 h after CFA injection (ipsilateral vs. contralateral: 12.3 ± 4.6 vs. 1.3 ± 1.9, P < 0.0001) and maintained at 24 h (10.0 ± 4.1 vs. 0.8 ± 1.3, P < 0.0001) (n = 4 to 6). CONCLUSION These results suggest that contribution of the αCGRP system depends on the modality of pain and the stage of inflammation.
Collapse
|
24
|
Reaction time and replenishment time of SP and CGRP after incision in rat skin. Open Life Sci 2014. [DOI: 10.2478/s11535-014-0349-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractBackground. The skin neurogenic inflammation is mainly related to Substance P (SP) and Calcitonin Gene-related Peptide (CGRP). There is no data on their availability in the dynamics of skin nerve endings, concerning their release and replenishment after a nociceptive stimulus, so this was investigated. Materials and methods. 25 rats were randomly distributed in 5 groups. The animals of the control group (CG) determined the baseline levels of neuropeptides in the skin. The groups S0 and S30 did not receive any cutaneous stimulus at 30 and 60 minutes, respectively. In the group S1, an “incision stimulus” was made at 30 minutes. In the group S31, a nociceptive stimulus was performed by subdermal scratching at 30 minutes and, at 60 minutes, the “incision stimulus” was carried out in the same location (“nociceptive hyperstimulation”). The skin samples of the other animals were harvested from the back 1 minute after their death. SP, pro-CGRP and CGRP were quantified by Western Blotting. Results. The “incision stimulus” released SP, S1 compared to S0 (p <0.05) detected in the first minute, and the replenishment time was more than 30 minutes. Also, it cleaved pro-CGRP, S1 compared to S31 (p <0.05) in the first minute, and its replenishment time less than 30 minutes. Release of CGRP was not detected. Conclusion. The incision released SP already detected in the first minute; its replenishment time is more than 30 minutes. The incision decreased pro-CGRP, also detected in the first minute; and its replenishment time is less than 30 minutes.
Collapse
|
25
|
Peripheral TGF-β1 signaling is a critical event in bone cancer-induced hyperalgesia in rodents. J Neurosci 2014; 33:19099-111. [PMID: 24305807 DOI: 10.1523/jneurosci.4852-12.2013] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pain is the most common symptom of bone cancer. TGF-β, a major bone-derived growth factor, is largely released by osteoclast bone resorption during the progression of bone cancer and contributes to proliferation, angiogenesis, immunosuppression, invasion, and metastasis. Here, we further show that TGF-β1 is critical for bone cancer-induced pain sensitization. We found that, after the progression of bone cancer, TGF-β1 was highly expressed in tumor-bearing bone, and the expression of its receptors, TGFβRI and TGFβRII, was significantly increased in the DRG in a rat model of bone cancer pain that is based on intratibia inoculation of Walker 256 mammary gland carcinoma cells. The blockade of TGF-β receptors by the TGFβRI antagonist SD-208 robustly suppressed bone cancer-induced thermal hyperalgesia on post-tumor day 14 (PTD 14). Peripheral injection of TGF-β1 directly induced thermal hyperalgesia in intact rats and wide-type mice, but not in Trpv1(-/-) mice. Whole-cell patch-clamp recordings from DRG neurons showed that transient receptor potential vanilloid (TRPV1) sensitivity was significantly enhanced on PTD 14. Extracellular application of TGF-β1 significantly potentiated TRPV1 currents and increased [Ca(2+)]i in DRG neurons. Pharmacological studies revealed that the TGF-β1 sensitization of TRPV1 and the induction of thermal hyperalgesia required the TGF-βR-mediated Smad-independent PKCε and TGF-β activating kinase 1-p38 pathways. These findings suggest that TGF-β1 signaling contributes to bone cancer pain via the upregulation and sensitization of TRPV1 in primary sensory neurons and that therapeutic targeting of TGF-β1 may ameliorate the bone cancer pain in advanced cancer.
Collapse
|
26
|
Guo H, Shen X, Xu Y, He Y, Hu W. The effect of activin A on signal transduction pathways in PC12 cells subjected to oxygen and glucose deprivation. Int J Mol Med 2013; 33:135-41. [PMID: 24173551 DOI: 10.3892/ijmm.2013.1539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 10/02/2013] [Indexed: 11/05/2022] Open
Abstract
The processes and mechanisms underlying brain injuries due to ischemia and anoxia have yet to be determined. Additionally, few clinical treatements are currently available. Activins have a protective role in the restoration, differentiation, and survival of injured cells, including Activin A (ActA), which acts as a neuroprotectant. However, its exact mechanism of action remains to be determined. ActA has been shown to protect neurons following ischemic brain injury. In this study, PC12 cells were differentiated into neuron-like cells after stimulation with nerve growth factor to prepare an oxygen/glucose deprivation (OGD) model in neurons. The differentiated PC12 cells, subjected to the OGD model, were exposed to ActA. Results showed that the PC12 survival rate decreased after OGD, leading to an increase in caspase-3 expression in these cells. Pretreatment with ActA was able to partially prevent OGD-induced apoptosis, likely through the downregulation of caspase-3. Futhermore, ActA pretreatment increased the expression of key proteins in the ActA/Smads signal transduction pathway, which may promote neuroprotection after OGD. Therefore, exogenous ActA may function as a neuroprotectant and provide a novel therapeutic treatment for ischemic brain injury.
Collapse
Affiliation(s)
- Hongliang Guo
- Department of Neurology, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100020, P.R. China
| | | | | | | | | |
Collapse
|
27
|
Activin A: A Potential Therapeutic Target for Characterizing and Stopping Joint Pain Early in Rheumatoid Arthritis Patients. Inflammation 2013; 37:170-6. [DOI: 10.1007/s10753-013-9727-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
28
|
Moura J, da Silva L, Cruz MT, Carvalho E. Molecular and cellular mechanisms of bone morphogenetic proteins and activins in the skin: potential benefits for wound healing. Arch Dermatol Res 2013; 305:557-69. [PMID: 23800970 DOI: 10.1007/s00403-013-1381-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 06/05/2013] [Accepted: 06/14/2013] [Indexed: 01/13/2023]
Abstract
Bone morphogenetic proteins (BMPs) and activins are phylogenetically conserved proteins, belonging to the transforming growth factor-β superfamily, that signal through the phosphorylation of receptor-regulated Smad proteins, activating different cell responses. They are involved in various steps of skin morphogenesis and wound repair, as can be evidenced by the fact that their expression is increased in skin injuries. BMPs play not only a role in bone regeneration but are also involved in cartilage, tendon-like tissue and epithelial regeneration, maintain vascular integrity, capillary sprouting, proliferation/migration of endothelial cells and angiogenesis, promote neuron and dendrite formation, alter neuropeptide levels and are involved in immune response modulation, at least in animal models. On the other hand, activins are involved in wound repair through the regulation of skin and immune cell migration and differentiation, re-epithelialization and granulation tissue formation, and also promote the expression of collagens by fibroblasts and modulate scar formation. This review aims at enunciating the effects of BMPs and activins in the skin, namely in skin development, as well as in crucial phases of skin wound healing, such as inflammation, angiogenesis and repair, and will focus on the effects of these proteins on skin cells and their signaling pathways, exploring the potential therapeutic approach of the application of BMP-2, BMP-6 and activin A in chronic wounds, particularly diabetic foot ulcerations.
Collapse
Affiliation(s)
- J Moura
- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | | | | | | |
Collapse
|
29
|
Porseva VV, Strelkov AA, Shilkin VV, Maslyukov PM. Age-related changes in sensory neurons containing calcitonin gene-related peptide under conditions of afferentation deficit in rats. Russ J Dev Biol 2012. [DOI: 10.1134/s1062360412060069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Zhu Y, Colak T, Shenoy M, Liu L, Mehta K, Pai R, Zou B, Xie XS, Pasricha PJ. Transforming growth factor beta induces sensory neuronal hyperexcitability, and contributes to pancreatic pain and hyperalgesia in rats with chronic pancreatitis. Mol Pain 2012; 8:65. [PMID: 22963239 PMCID: PMC3515355 DOI: 10.1186/1744-8069-8-65] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 09/07/2012] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Transforming growth factor beta (TGFβ) is upregulated in chronic inflammation, where it plays a key role in wound healing and promoting fibrosis. However, little is known about the peripheral effects of TGFβ on nociception. METHODS We tested the in vitro effects of TGFβ1 on the excitability of dorsal root ganglia (DRG) neurons and the function of potassium (K) channels. We also studied the effects of TGFβ1 infusion on pain responses to noxious electrical stimulation in healthy rats as well as the effects of neutralization of TGFβ1 on evoked pain behaviors in a rat model of chronic pancreatitis. RESULTS Exposure to TGFβ1 in vitro increased sensory neuronal excitability, decreased voltage-gated A-type K(+) currents (IA) and downregulated expression of the Kv1.4 (KCNA4) gene. Further TGFβ1 infusion into the naïve rat pancreas in vivo induces hyperalgesia and conversely, neutralization of TGFβ1 attenuates hyperalgesia only in rats with experimental chronic pancreatitis. Paradoxically, TGFβ1 neutralization in naïve rats results in pancreatic hyperalgesia. CONCLUSIONS TGFβ1 is an important and complex modulator of sensory neuronal function in chronic inflammation, providing a link between fibrosis and nociception and is a potentially novel target for the treatment of persistent pain associated with chronic pancreatitis.
Collapse
Affiliation(s)
- Yaohui Zhu
- Johns Hopkins Center for Neurogastroenterology, Department of Medicine, Division of Gastroenterology and Hepatology, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Yu SJ, Xia CM, Kay JC, Qiao LY. Activation of extracellular signal-regulated protein kinase 5 is essential for cystitis- and nerve growth factor-induced calcitonin gene-related peptide expression in sensory neurons. Mol Pain 2012; 8:48. [PMID: 22742729 PMCID: PMC3502118 DOI: 10.1186/1744-8069-8-48] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 06/24/2012] [Indexed: 11/23/2022] Open
Abstract
Background Cystitis causes considerable neuronal plasticity in the primary afferent pathways. The molecular mechanism and signal transduction underlying cross talk between the inflamed urinary bladder and sensory sensitization has not been investigated. Results In a rat cystitis model induced by cyclophosphamide (CYP) for 48 h, the mRNA and protein levels of the excitatory neurotransmitter calcitonin gene-related peptide (CGRP) are increased in the L6 dorsal root ganglia (DRG) in response to bladder inflammation. Cystitis-induced CGRP expression in L6 DRG is triggered by endogenous nerve growth factor (NGF) because neutralization of NGF with a specific NGF antibody reverses CGRP up-regulation during cystitis. CGRP expression in the L6 DRG neurons is also enhanced by retrograde NGF signaling when NGF is applied to the nerve terminals of the ganglion-nerve two-compartmented preparation. Characterization of the signaling pathways in cystitis- or NGF-induced CGRP expression reveals that the activation (phosphorylation) of extracellular signal-regulated protein kinase (ERK)5 but not Akt is involved. In L6 DRG during cystitis, CGRP is co-localized with phospho-ERK5 but not phospho-Akt. NGF-evoked CGRP up-regulation is also blocked by inhibition of the MEK/ERK pathway with specific MEK inhibitors U0126 and PD98059, but not by inhibition of the PI3K/Akt pathway with inhibitor LY294002. Further examination shows that cystitis-induced cAMP-responsive element binding protein (CREB) activity is expressed in CGRP bladder afferent neurons and is co-localized with phospho-ERK5 but not phospho-Akt. Blockade of NGF action in vivo reduces the number of DRG neurons co-expressing CGRP and phospho-CREB, and reverses cystitis-induced increases in micturition frequency. Conclusions A specific pathway involving NGF-ERK5-CREB axis plays an essential role in cystitis-induced sensory activation.
Collapse
Affiliation(s)
- Sharon J Yu
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | | | | | | |
Collapse
|
32
|
Utreras E, Keller J, Terse A, Prochazkova M, Iadarola MJ, Kulkarni AB. Transforming growth factor-β1 regulates Cdk5 activity in primary sensory neurons. J Biol Chem 2012; 287:16917-29. [PMID: 22451679 DOI: 10.1074/jbc.m111.329979] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In addition to many important roles for Cdk5 in brain development and synaptic function, we reported previously that Cdk5 regulates inflammatory pain signaling, partly through phosphorylation of transient receptor potential vanilloid 1 (TRPV1), an important Na(+)/Ca(2+) channel expressed in primary nociceptive afferent nerves. Because TGF-β regulates inflammatory processes and its receptor is expressed in TRPV1-positive afferents, we studied the cross-talk between these two pathways in sensory neurons during experimental peripheral inflammation. We demonstrate that TGF-β1 increases transcription and protein levels of the Cdk5 co-activator p35 through ERK1/2, resulting in an increase in Cdk5 activity in rat B104 neuroblastoma cells. Additionally, TGF-β1 enhances the capsaicin-induced Ca(2+) influx in cultured primary neurons from dorsal root ganglia (DRG). Importantly, Cdk5 activity was reduced in the trigeminal ganglia and DRG of 14-day-old TGF-β1 knock-out mice, resulting in reduced Cdk5-dependent phosphorylation of TRPV1. The decreased Cdk5 activity is associated with attenuated thermal hyperalgesia in TGF-β1 receptor conditional knock-out mice, where TGF-β signaling is significantly reduced in trigeminal ganglia and DRG. Collectively, our results indicate that active cross-talk between the TGF-β and Cdk5 pathways contributes to inflammatory pain signaling.
Collapse
Affiliation(s)
- Elias Utreras
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, NIDCR, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
33
|
Eade KT, Fancher HA, Ridyard MS, Allan DW. Developmental transcriptional networks are required to maintain neuronal subtype identity in the mature nervous system. PLoS Genet 2012; 8:e1002501. [PMID: 22383890 PMCID: PMC3285578 DOI: 10.1371/journal.pgen.1002501] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 12/09/2011] [Indexed: 11/30/2022] Open
Abstract
During neurogenesis, transcription factors combinatorially specify neuronal fates and then differentiate subtype identities by inducing subtype-specific gene expression profiles. But how is neuronal subtype identity maintained in mature neurons? Modeling this question in two Drosophila neuronal subtypes (Tv1 and Tv4), we test whether the subtype transcription factor networks that direct differentiation during development are required persistently for long-term maintenance of subtype identity. By conditional transcription factor knockdown in adult Tv neurons after normal development, we find that most transcription factors within the Tv1/Tv4 subtype transcription networks are indeed required to maintain Tv1/Tv4 subtype-specific gene expression in adults. Thus, gene expression profiles are not simply “locked-in,” but must be actively maintained by persistent developmental transcription factor networks. We also examined the cross-regulatory relationships between all transcription factors that persisted in adult Tv1/Tv4 neurons. We show that certain critical cross-regulatory relationships that had existed between these transcription factors during development were no longer present in the mature adult neuron. This points to key differences between developmental and maintenance transcriptional regulatory networks in individual neurons. Together, our results provide novel insight showing that the maintenance of subtype identity is an active process underpinned by persistently active, combinatorially-acting, developmental transcription factors. These findings have implications for understanding the maintenance of all long-lived cell types and the functional degeneration of neurons in the aging brain. For neurons to function properly, they must establish and then maintain their unique, subtype-specific gene expression profiles. These unique gene expression profiles are established during development by networks of DNA–binding proteins, termed transcription factors (TFs). However, how neurons maintain their unique gene expression profiles in the mature and aging brain is largely unknown. Recent advances in inducible genetic technologies now allow us to manipulate gene expression in adult neurons, after normal development. Applying such techniques, we examined the effect of knocking down TF expression in two adult neuronal subtypes. We show that the TF networks that establish unique gene expression profiles during development are then required to maintain them thereafter. Thus, gene expression profiles are not simply “locked-in,” but must be actively maintained by persistent developmental TF networks. However, we found that critical cross-regulatory relationships that had existed between TFs during development were not present in the adult, even between persisting TFs. This highlights important differences between developmental and maintenance transcriptional networks in individual neurons. The dependence of subtype gene expression on active mechanisms represents a potential Achilles heel for long-lived cells, as deterioration of those active mechanisms could lead to functional degeneration of neurons with advancing age.
Collapse
Affiliation(s)
- Kevin T. Eade
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Hailey A. Fancher
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Marc S. Ridyard
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Douglas W. Allan
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
- * E-mail:
| |
Collapse
|
34
|
Liu XJ, Zhang FX, Liu H, Li KC, Lu YJ, Wu QF, Li JY, Wang B, Wang Q, Lin LB, Zhong YQ, Xiao HS, Bao L, Zhang X. Activin C expressed in nociceptive afferent neurons is required for suppressing inflammatory pain. Brain 2012; 135:391-403. [DOI: 10.1093/brain/awr350] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Xing-Jun Liu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fang-Xiong Zhang
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Liu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kai-Cheng Li
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ying-Jin Lu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qing-Feng Wu
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jia-Yin Li
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Wang
- 2 State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiong Wang
- 2 State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li-Bo Lin
- 3 National Engineering Centre for Biochip at Shanghai, Shanghai 201203, China
| | - Yan-Qing Zhong
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hua-Sheng Xiao
- 3 National Engineering Centre for Biochip at Shanghai, Shanghai 201203, China
| | - Lan Bao
- 2 State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xu Zhang
- 1 State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
35
|
Transforming growth factor-β in normal nociceptive processing and pathological pain models. Mol Neurobiol 2011; 45:76-86. [PMID: 22125199 DOI: 10.1007/s12035-011-8221-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2011] [Accepted: 11/09/2011] [Indexed: 12/20/2022]
Abstract
The transforming growth factor-β (TGF-β) superfamily is a multifunctional, contextually acting family of cytokines that participate in the regulation of development, disease and tissue repair in the nervous system. The TGF-β family is composed of several members, including TGF-βs, bone morphogenetic proteins (BMPs) and activins. In this review, we discuss recent findings that suggest TGF-β function as important pleiotropic modulators of nociceptive processing both physiologically and under pathological painful conditions. The strategy of increasing TGF-β signaling by deleting "BMP and activin membrane-bound inhibitor" (BAMBI), a TGF-β pseudoreceptor, has demonstrated the inhibitory role of TGF-β signaling pathways in normal nociception and in inflammatory and neuropathic pain models. In particular, strong evidence suggests that TGF-β1 is a relevant mediator of nociception and has protective effects against the development of chronic neuropathic pain by inhibiting the neuroimmune responses of neurons and glia and promoting the expression of endogenous opioids within the spinal cord. In the peripheral nervous system, activins and BMPs function as target-derived differentiation factors that determine and maintain the phenotypic identity and circuit assembly of peptidergic nociceptors. In this context, activin is involved in the complex events of neuroinflammation that modulate the expression of pain during wound healing. These findings have provided new insights into the physiopathology of nociception. Moreover, specific members of the TGF-β family and their signaling effectors and modulator molecules may be promising molecular targets for novel therapeutic agents for pain management.
Collapse
|
36
|
Wang Z, Chabot JG, Quirion R. On the possible role of ERK, p38 and CaMKII in the regulation of CGRP expression in morphine-tolerant rats. Mol Pain 2011; 7:68. [PMID: 21933441 PMCID: PMC3190348 DOI: 10.1186/1744-8069-7-68] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 09/21/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The neuropeptide, calcitonin gene-related peptide (CGRP) has been proposed to be a regulator of the development of morphine analgesic tolerance and thereby could be a target to reduce the induction of this phenomenon under clinical conditions. However, the mechanisms of CGRP regulation are unclear. We investigated here the possible role of the extracellular signal-regulated protein kinase (ERK), p38 and calcium/calmodulin-dependent protein kinase II (CaMKII) in CGRP regulation following chronic morphine treatment. RESULTS A 7-day treatment with morphine (15 μg/day) led to an increase in CGRP contents in the spinal cord dorsal horn (SCDH) and dorsal root ganglion (DRG) and this effect was prevented by the inhibition of the ERK, p38 or CaMKII pathway. The phosphorylation/activation of ERK, p38 and CaMKII was enhanced in the SCDH following chronic morphine while in DRG only the phosphorylation of CaMKII was increased. Moreover, our chronic morphine treatment up-regulated neuronal nitric oxide synthase (nNOS) levels in the SCDH, an effect blocked by the inhibition of the ERK, p38 or CaMKII pathway. The blockade of nNOS activity also suppressed chronic morphine-induced CGRP increases in the DRG and SCDH. Double immunofluorescence studies revealed that nNOS and CaMKII are co-localized in the SCDH and that CaMKII is activated in CGRP-expressing DRG neurons. CONCLUSIONS The activation of spinal ERK, p38 and CaMKII, alongside nNOS, is involved in chronic morphine-induced CGRP up-regulation in both the DRG and SCDH. Moreover, the stimulation of CaMKII in the DRG likely directly regulates the expression of CGRP associated with morphine analgesic tolerance.
Collapse
Affiliation(s)
- Zhiyong Wang
- Dept, of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H 1R3, Canada
| | | | | |
Collapse
|
37
|
Mishima T, Ito Y, Hosono K, Tamura Y, Uchida Y, Hirata M, Suzsuki T, Amano H, Kato S, Kurihara Y, Kurihara H, Hayashi I, Watanabe M, Majima M. Calcitonin gene-related peptide facilitates revascularization during hindlimb ischemia in mice. Am J Physiol Heart Circ Physiol 2011; 300:H431-9. [DOI: 10.1152/ajpheart.00466.2010] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It is known that the neural system plays a fundamental role in neovascularization. A neuropeptide, calcitonin gene-related peptide (CGRP), is widely distributed in the central and peripheral neuronal systems. However, it remains to be elucidated the role of CGRP in angiogenesis during ischemia. The present study examined whether endogenous CGRP released from neuronal systems facilitates revascularization in response to ischemia using CGRP knockout mice (CGRP−/−). CGRP−/− or their wild-type littermates (CGRP+/+) were subjected to unilateral hindlimb ischemia. CGRP−/− exhibited impaired blood flow recovery from ischemia and decreased capillary density expressed in terms of the number of CD-31-positive cells in the ischemic tissues compared with CGRP+/+. In vivo microscopic studies showed that the functional capillary density in CGRP−/− was reduced. Hindlimb ischemia increased the expression of pro-CGRP mRNA and of CGRP protein in the lumbar dorsal root ganglia. Lack of CGRP decreased mRNA expression of growth factors, including CD31, vascular endothelial growth factor-A, basic fibroblast growth factor, and transforming growth factor-β, in the ischemic limb tissue. The application of CGRP enhanced the mRNA expression of CD31 and VEGF-A in human umbilical vein endothelial cells (HUVECs) and fibroblasts. Subcutaneous infusion of CGRP8–37, a CGRP antagonist, using miniosmotic pumps delayed angiogenesis and reduced the expression of proangiogenic growth factors during hindlimb ischemia. These results indicate that endogenous CGRP facilitates angiogenesis in response to ischemia. Targeting CGRP may provide a promising approach for controlling angiogenesis related to pathophysiological conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Shintaro Kato
- Cardio-angiology, Kitasato University School of Medicine, Kanagawa
| | - Yukiko Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, Tokyo University, Tokyo; and
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, Tokyo University, Tokyo; and
| | - Izumi Hayashi
- Faculty of Pharmaceutical Sciences, Department of Pathophysiology, Nippon Pharmaceutical University, Saitama, Japan
| | | | | |
Collapse
|
38
|
da Silva S, Wang F. Retrograde neural circuit specification by target-derived neurotrophins and growth factors. Curr Opin Neurobiol 2010; 21:61-7. [PMID: 20810276 DOI: 10.1016/j.conb.2010.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 07/15/2010] [Accepted: 07/21/2010] [Indexed: 01/13/2023]
Abstract
Neural circuit assembly during development involves a series of highly regulated steps. While genetically pre-determined programs play key roles in the early steps including neurogenesis, migration, and initial growth and guidance of axons; increasing evidence indicates that as the axons reach their targets, the late steps of neuronal differentiation and connectivity formation may be influenced or even specified by target-derived signals. Here we attempt to provide a brief synthesized review on the roles of retrograde neurotrophin and growth factor signaling in regulating the final stages of neural circuit specificity such as axonal projection, dendritic patterning, neurotransmitter phenotype acquisition, and synapse formation.
Collapse
Affiliation(s)
- Susana da Silva
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC 27710, USA
| | | |
Collapse
|
39
|
BAMBI (bone morphogenetic protein and activin membrane-bound inhibitor) reveals the involvement of the transforming growth factor-beta family in pain modulation. J Neurosci 2010; 30:1502-11. [PMID: 20107078 DOI: 10.1523/jneurosci.2584-09.2010] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Transforming growth factors-beta (TGF-betas) signal through type I and type II serine-threonine kinase receptor complexes. During ligand binding, type II receptors recruit and phosphorylate type I receptors, triggering downstream signaling. BAMBI [bone morphogenetic protein (BMP) and activin membrane-bound inhibitor] is a transmembrane pseudoreceptor structurally similar to type I receptors but lacks the intracellular kinase domain. BAMBI modulates negatively pan-TGF-beta family signaling; therefore, it can be used as an instrument for unraveling the roles of these cytokines in the adult CNS. BAMBI is expressed in regions of the CNS involved in pain transmission and modulation. The lack of BAMBI in mutant mice resulted in increased levels of TGF-beta signaling activity, which was associated with attenuation of acute pain behaviors, regardless of the modality of the stimuli (thermal, mechanical, chemical/inflammatory). The nociceptive hyposensitivity exhibited by BAMBI(-/-) mice was reversed by the opioid antagonist naloxone. Moreover, in a model of chronic neuropathic pain, the allodynic responses of BAMBI(-/-) mice also appeared attenuated through a mechanism involving delta-opioid receptor signaling. Basal mRNA and protein levels of precursor proteins of the endogenous opioid peptides proopiomelanocortin (POMC) and proenkephalin (PENK) appeared increased in the spinal cords of BAMBI(-/-). Transcript levels of TGF-betas and their intracellular effectors correlated directly with genes encoding opioid peptides, whereas BAMBI correlated inversely. Furthermore, incubation of spinal cord explants with activin A or BMP-7 increased POMC and/or PENK mRNA levels. Our findings identify TGF-beta family members as modulators of acute and chronic pain perception through the transcriptional regulation of genes encoding the endogenous opioids.
Collapse
|
40
|
Bruna J, Udina E, Alé A, Vilches JJ, Vynckier A, Monbaliu J, Silverman L, Navarro X. Neurophysiological, histological and immunohistochemical characterization of bortezomib-induced neuropathy in mice. Exp Neurol 2010; 223:599-608. [PMID: 20188093 DOI: 10.1016/j.expneurol.2010.02.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 02/09/2010] [Accepted: 02/13/2010] [Indexed: 12/11/2022]
Abstract
Bortezomib, a proteasome inhibitor, is an antineoplastic drug to treat multiple myeloma and mantle cell lymphoma. Its most clinically significant adverse event is peripheral sensory neuropathy. Our objective was to characterize the neuropathy induced by bortezomib in a mouse model. Two groups were used; one group received vehicle solution and another bortezomib (1mg/kg/twice/week) for 6weeks (total dose as human schedule). Tests were performed during treatment and for 4weeks post dosing to evaluate electrophysiological, autonomic, pain sensibility and sensory-motor function changes. At the end of treatment and after washout, sciatic and tibial nerves, dorsal ganglia and intraepidermal innervation were analyzed. Bortezomib induced progressive significant decrease of sensory action potential amplitude, mild reduction of sensory velocities without effect in motor conductions. Moreover, it significantly increased pain threshold and sensory-motor impairment at 6weeks. According to these data, histopathological findings shown a mild reduction of myelinated (-10%; p=0.001) and unmyelinated fibers (-27%; p=0.04), mostly involving large and C fibers, with abnormal vesicular inclusion body in unmyelinated axons. Neurons were also involved as shown by immunohistochemical phenotypic switch. After washout, partial recovery was observed in functional, electrophysiological and histological analyses. These results suggest that axon and myelin changes might be secondary to an initial dysfunctional neuronopathy.
Collapse
Affiliation(s)
- Jordi Bruna
- Group of Neuroplasticity and Regeneration, Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Neuronal phenotype in the mature nervous system is maintained by persistent retrograde bone morphogenetic protein signaling. J Neurosci 2009; 29:3852-64. [PMID: 19321782 DOI: 10.1523/jneurosci.0213-09.2009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The terminal differentiation of many developing neurons occurs after they innervate their target cells and is triggered by secreted target-derived signals that are transduced by presynaptic cognate receptors. Such retrograde signaling induces the expression of genes that are often distinctive markers of neuronal phenotype and function. However, whether long-term maintenance of neuronal phenotype requires persistent retrograde signaling remains poorly understood. Previously, we demonstrated that retrograde bone morphogenetic protein (BMP) signaling induces expression of a phenotypic marker of Drosophila Tv neurons, the neuropeptide FMRFamide (FMRFa). Here, we used a genetic technique that spatiotemporally targets transgene expression in Drosophila to test the role of persistent BMP signaling in the maintenance of Tv phenotype. We show that expression of dominant blockers of BMP signaling selectively in adult Tv neurons dramatically downregulated FMRFa expression. Moreover, adult-onset expression of mutant Glued, which blocks dynein/dynactin-mediated retrograde axonal transport, eliminated retrograde BMP signaling and dramatically downregulated FMRFa expression. Finally, we found that BMP deprivation did not affect Tv neuron survival and that FMRFa expression fully recovered to control levels after the termination of BMP blockade or Glued expression. Our results show that persistent retrograde BMP signaling is required to induce and to subsequently maintain the expression of a stably expressed phenotypic marker in a subset of mature Drosophila neurons. We postulate that retrograde maintenance of neuronal phenotype is conserved in vertebrates, and as a consequence, neuronal phenotype is likely vulnerable to neurodegenerative disease pathologies that disrupt neuronal connectivity or axonal transport.
Collapse
|
42
|
Uchida H, Matsumoto M, Ueda H. Profiling of BoNT/C3-reversible gene expression induced by lysophosphatidic acid: ephrinB1 gene up-regulation underlying neuropathic hyperalgesia and allodynia. Neurochem Int 2008; 54:215-21. [PMID: 19111589 DOI: 10.1016/j.neuint.2008.11.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 10/27/2008] [Accepted: 11/10/2008] [Indexed: 12/11/2022]
Abstract
Lysophosphatidic acid (LPA) signaling, through LPA(1) receptor and its downstream RhoA, has been reported to initiate nerve injury-induced neuropathic pain. In the present study, we performed gene expression profiling of the dorsal root ganglion (DRG) to identify genes induced by intrathecal injection of LPA in a botulinum toxin C3 (BoNT/C3)-reversible manner. We selected and functionally characterized ephrinB1 from 82 identified genes as a potential gene involved in pain transmission, since ephrinB1 is implicated to modulate N-methyl-d-aspartate (NMDA) receptor functions in spinal pain transmission. The LPA-induced and BoNT/C3-reversible ephrinB1 gene expression was confirmed by quantitative real-time PCR. Furthermore, treatments with an antisense oligodeoxynucleotide for ephrinB1 largely abolished the LPA-induced thermal hyperalgesia and allodynia in response to mechanical or Abeta-fiber-mediated electrical stimuli on day 1 after the injection. In addition, intrathecal treatment with a soluble ligand, ephrinB1-Fc, caused similar neuropathic pain-like behaviors in a manner that was reversible by the NMDA receptor antagonist MK-801. These results suggest that ephrinB1 plays a crucial role in LPA-induced neuropathic pain. In addition, the present study may provide a new strategy to identify unique neuropathic pain-related genes.
Collapse
Affiliation(s)
- Hitoshi Uchida
- Nagasaki University Graduate School of Biomedical Sciences, Bunkyo-machi, Japan
| | | | | |
Collapse
|
43
|
Toda M, Suzuki T, Hosono K, Hayashi I, Hashiba S, Onuma Y, Amano H, Kurihara Y, Kurihara H, Okamoto H, Hoka S, Majima M. Neuronal system-dependent facilitation of tumor angiogenesis and tumor growth by calcitonin gene-related peptide. Proc Natl Acad Sci U S A 2008; 105:13550-5. [PMID: 18757746 PMCID: PMC2527353 DOI: 10.1073/pnas.0800767105] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Indexed: 11/18/2022] Open
Abstract
A neuropeptide, calcitonin gene-related peptide (CGRP), is widely distributed in neuronal systems and exhibits numerous biological activities. Using CGRP-knockout mice (CGRP(-/-)), we examined whether or not endogenous CGRP facilitates angiogenesis indispensable to tumor growth. CGRP increased tube formation by endothelial cells in vitro and enhanced sponge-induced angiogenesis in vivo. Tumor growth and tumor-associated angiogenesis in CGRP(-/-) implanted with Lewis lung carcinoma (LLC) cells were significantly reduced compared with those in wild-type (WT) mice. A CGRP antagonist, CGRP8-37 or denervation of sciatic nerves (L(1-5)) suppressed LLC growth in the sites of denervation compared with vehicle infusion or sham operation. CGRP precursor mRNA levels in the dorsal root ganglion in LLC-bearing WT were increased compared with those in non-LLC-bearing mice. This increase was abolished by denervation. The expression of VEGF in tumor stroma was down-regulated in CGRP(-/-). These results indicate that endogenous CGRP facilitates tumor-associated angiogenesis and tumor growth and suggest that relevant CGRP may be derived from neuronal systems including primary sensory neurons and may become a therapeutic target for cancers.
Collapse
Affiliation(s)
- Masaya Toda
- Departments of *Pharmacology and
- Anesthesiology, Kitasato University School of Medicine, Kanagawa 228-8555, Japan
| | | | | | - Izumi Hayashi
- Department of Pathophysiology, Faculty of Pharmaceutical Sciences, Nippon Pharmaceutical University, Saitama 362-0806, Japan; and
| | - Shinichiro Hashiba
- Anesthesiology, Kitasato University School of Medicine, Kanagawa 228-8555, Japan
| | | | | | - Yukiko Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, Tokyo University, Tokyo 113-0033, Japan
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, Tokyo University, Tokyo 113-0033, Japan
| | - Hirotsugu Okamoto
- Anesthesiology, Kitasato University School of Medicine, Kanagawa 228-8555, Japan
| | - Sumio Hoka
- Anesthesiology, Kitasato University School of Medicine, Kanagawa 228-8555, Japan
| | | |
Collapse
|
44
|
Modulation of chloride homeostasis by inflammatory mediators in dorsal root ganglion neurons. Mol Pain 2008; 4:32. [PMID: 18700020 PMCID: PMC2526990 DOI: 10.1186/1744-8069-4-32] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Accepted: 08/12/2008] [Indexed: 01/10/2023] Open
Abstract
Background Chloride currents in peripheral nociceptive neurons have been implicated in the generation of afferent nociceptive signals, as Cl- accumulation in sensory endings establishes the driving force for depolarizing, and even excitatory, Cl- currents. The intracellular Cl- concentration can, however, vary considerably between individual DRG neurons. This raises the question, whether the contribution of Cl- currents to signal generation differs between individual afferent neurons, and whether the specific Cl- levels in these neurons are subject to modulation. Based on the hypothesis that modulation of the peripheral Cl- homeostasis is involved in the generation of inflammatory hyperalgesia, we examined the effects of inflammatory mediators on intracellular Cl- concentrations and on the expression levels of Cl- transporters in rat DRG neurons. Results We developed an in vitro assay for testing how inflammatory mediators influence Cl- concentration and the expression of Cl- transporters. Intact DRGs were treated with 100 ng/ml NGF, 1.8 μM ATP, 0.9 μM bradykinin, and 1.4 μM PGE2 for 1–3 hours. Two-photon fluorescence lifetime imaging with the Cl--sensitive dye MQAE revealed an increase of the intracellular Cl- concentration within 2 hours of treatment. This effect coincided with enhanced phosphorylation of the Na+-K+-2Cl- cotransporter NKCC1, suggesting that an increased activity of that transporter caused the early rise of intracellular Cl- levels. Immunohistochemistry of NKCC1 and KCC2, the main neuronal Cl- importer and exporter, respectively, exposed an inverse regulation by the inflammatory mediators. While the NKCC1 immunosignal increased, that of KCC2 declined after 3 hours of treatment. In contrast, the mRNA levels of the two transporters did not change markedly during this time. These data demonstrate a fundamental transition in Cl- homeostasis toward a state of augmented Cl- accumulation, which is induced by a 1–3 hour treatment with inflammatory mediators. Conclusion Our findings indicate that inflammatory mediators impact on Cl- homeostasis in DRG neurons. Inflammatory mediators raise intracellular Cl- levels and, hence, the driving force for depolarizing Cl- efflux. These findings corroborate current concepts for the role of Cl- regulation in the generation of inflammatory hyperalgesia and allodynia. As the intracellular Cl- concentration rises in DRG neurons, afferent signals can be boosted by excitatory Cl- currents in the presynaptic terminals. Moreover, excitatory Cl- currents in peripheral sensory endings may also contribute to the generation or modulation of afferent signals, especially in inflamed tissue.
Collapse
|
45
|
Young KG, Kothary R. Dystonin/Bpag1 is a necessary endoplasmic reticulum/nuclear envelope protein in sensory neurons. Exp Cell Res 2008; 314:2750-61. [PMID: 18638474 DOI: 10.1016/j.yexcr.2008.06.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 06/24/2008] [Accepted: 06/25/2008] [Indexed: 11/17/2022]
Abstract
Dystonin/Bpag1 proteins are cytoskeletal linkers whose loss of function in mice results in a hereditary sensory neuropathy with a progressive loss of limb coordination starting in the second week of life. These mice, named dystonia musculorum (dt), succumb to the disease and die of unknown causes prior to sexual maturity. Previous evidence indicated that cytoskeletal defects in the axon are a primary cause of dt neurodegeneration. However, more recent data suggests that other factors may be equally important contributors to the disease process. In the present study, we demonstrate perikaryal defects in dorsal root ganglion (DRG) neurons at stages preceding the onset of loss of limb coordination in dt mice. Abnormalities include alterations in endoplasmic reticulum (ER) chaperone protein expression, indicative of an ER stress response. Dystonin in sensory neurons localized in association with the ER and nuclear envelope (NE). A fusion protein ofthe dystonin-a2 isoform, which harbors an N-terminal transmembrane domain, associated with and reorganized the ER in cell culture. This isoform also interacts with the NE protein nesprin-3alpha, but not nesprin-3beta. Defects in dt mice, as demonstrated here, may ultimately result in pathogenesis involving ER dysfunction and contribute significantly to the dt phenotype.
Collapse
Affiliation(s)
- Kevin G Young
- Ottawa Health Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | | |
Collapse
|
46
|
Lu SG, Gold MS. Inflammation-induced increase in evoked calcium transients in subpopulations of rat dorsal root ganglion neurons. Neuroscience 2008; 153:279-88. [PMID: 18367340 PMCID: PMC2396945 DOI: 10.1016/j.neuroscience.2008.02.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Revised: 01/21/2008] [Accepted: 02/05/2008] [Indexed: 12/21/2022]
Abstract
The concentration of intracellular Ca(2+) ([Ca(2+)](i)) influences neuronal properties ranging from excitability to neurotransmitter release. Persistent inflammation is associated with changes in the properties of primary afferent neurons ranging from excitability to transmitter release. The purpose of the present study was to determine whether previously described inflammation-induced changes in excitability and transmitter release are associated with changes in the regulation of [Ca(2+)](i). Acutely dissociated dorsal root ganglion (DRG) neurons harvested from adult rats 3 days following a hind-paw injection of complete Freund's adjuvant (CFA) or naïve controls, were stimulated with 30 mM K(+) (High K(+)). High K(+) evoked changes in [Ca(2+)](i) were assessed with fura-2 ratiometric microfluorimetry. Subpopulations of DRG neurons were defined by cell body diameter, isolectin B4 (IB4) binding, capsaicin (CAP) sensitivity and target of innervation (1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbo-cyanine perchlorate labeling). Inflammation was associated with significant increases in resting [Ca(2+)](i) and increases in the magnitude and decreases in the decay, of the evoked increase in [Ca(2+)](i). The changes in evoked transients were larger in neurons innervating the site of inflammation. Furthermore, there were differences among subpopulations of DRG neurons with respect to changes in magnitude and/or decay of the evoked transient such that the increase in magnitude was larger in small- and medium-diameter neurons than in large diameter neurons while the decrease in the decay was greater in CAP responsive, IB4 positive, small- and medium-diameter neurons than in CAP unresponsive, IB4 negative and/or large-diameter neurons. These changes in the regulation of [Ca(2+)](i) were not due to inflammation-induced changes in passive or active electrophysiological properties. Importantly, an inflammation-induced increase in evoked Ca(2+) transients in putative nociceptive afferents may contribute to the pain and hyperalgesia associated with persistent inflammation via facilitation of transmitter release from these afferents.
Collapse
Affiliation(s)
- Shao-Gang Lu
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA 15213
| | - Michael S. Gold
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213
| |
Collapse
|
47
|
Bucelli RC, Gonsiorek EA, Kim WY, Bruun D, Rabin RA, Higgins D, Lein PJ. Statins decrease expression of the proinflammatory neuropeptides calcitonin gene-related peptide and substance P in sensory neurons. J Pharmacol Exp Ther 2007; 324:1172-80. [PMID: 18079356 DOI: 10.1124/jpet.107.132795] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Clinical and experimental observations suggest that statins may be useful for treating diseases presenting with predominant neurogenic inflammation, but the mechanism(s) mediating this potential therapeutic effect are poorly understood. In this study, we tested the hypothesis that statins act directly on sensory neurons to decrease expression of proinflammatory neuropeptides that trigger neurogenic inflammation, specifically calcitonin gene-related peptide (CGRP) and substance P. Reverse transcriptase-polymerase chain reaction, radioimmunoassay, and immunocytochemistry were used to quantify CGRP and substance P expression in dorsal root ganglia (DRG) harvested from adult male rats and in primary cultures of sensory neurons derived from embryonic rat DRG. Systemic administration of statins at pharmacologically relevant doses significantly reduced CGRP and substance P levels in DRG in vivo. In cultured sensory neurons, statins blocked bone morphogenetic protein (BMP)-induced CGRP and substance P expression and decreased expression of these neuropeptides in sensory neurons pretreated with BMPs. These effects were concentration-dependent and occurred independent of effects on cell survival or axon growth. Statin inhibition of neuropeptide expression was reversed by supplementation with mevalonate and cholesterol, but not isoprenoid precursors. BMPs signal via Smad activation, and cholesterol depletion by statins inhibited Smad1 phosphorylation and nuclear translocation. These findings identify a novel action of statins involving down-regulation of proinflammatory neuropeptide expression in sensory ganglia via cholesterol depletion and decreased Smad1 activation and suggest that statins may be effective in attenuating neurogenic inflammation.
Collapse
Affiliation(s)
- Robert C Bucelli
- Oregon Health Science University, CROET/L606, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Zhu W, Xu P, Cuascut FX, Hall AK, Oxford GS. Activin acutely sensitizes dorsal root ganglion neurons and induces hyperalgesia via PKC-mediated potentiation of transient receptor potential vanilloid I. J Neurosci 2007; 27:13770-80. [PMID: 18077689 PMCID: PMC6673610 DOI: 10.1523/jneurosci.3822-07.2007] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2007] [Revised: 09/19/2007] [Accepted: 09/19/2007] [Indexed: 01/13/2023] Open
Abstract
Pain hypersensitivity is a cardinal sign of tissue damage, but how molecules from peripheral tissues affect sensory neuron physiology is incompletely understood. Previous studies have shown that activin A increases after peripheral injury and is sufficient to induce acute nociceptive behavior and increase pain peptides in sensory ganglia. This study was designed to test the possibility that the enhanced nociceptive responsiveness associated with activin involved sensitization of transient receptor potential vanilloid I (TRPV1) in primary sensory neurons. Activin receptors were found widely distributed among adult sensory neurons, including those that also express the capsaicin receptor. Whole-cell patch-clamp recording from sensory neurons showed that activin acutely sensitized capsaicin responses and depended on activin receptor kinase activity. Pharmacological studies revealed that the activin sensitization of capsaicin responses required PKCepsilon signaling, but not PI3K (phosphoinositide 3-kinase), ERK (extracellular signal-regulated protein kinase), PKA, PKCalpha/beta, or Src. Furthermore, activin administration caused acute thermal hyperalgesia in wild-type mice, but not in TRPV1-null mice. These data suggest that activin signals through its own receptor, involves PKCepsilon signaling to sensitize the TRPV1 channel, and contributes to acute thermal hyperalgesia.
Collapse
Affiliation(s)
- Weiguo Zhu
- Stark Neurosciences Research Institute and Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, and
| | - Pin Xu
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio 44106
| | - Fernando X. Cuascut
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio 44106
| | - Alison K. Hall
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio 44106
| | - Gerry S. Oxford
- Stark Neurosciences Research Institute and Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, and
| |
Collapse
|
49
|
Hodge LK, Klassen MP, Han BX, Yiu G, Hurrell J, Howell A, Rousseau G, Lemaigre F, Tessier-Lavigne M, Wang F. Retrograde BMP signaling regulates trigeminal sensory neuron identities and the formation of precise face maps. Neuron 2007; 55:572-86. [PMID: 17698011 DOI: 10.1016/j.neuron.2007.07.010] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Revised: 05/31/2007] [Accepted: 07/12/2007] [Indexed: 01/22/2023]
Abstract
Somatosensory information from the face is transmitted to the brain by trigeminal sensory neurons. It was previously unknown whether neurons innervating distinct areas of the face possess molecular differences. We have identified a set of genes differentially expressed along the dorsoventral axis of the embryonic mouse trigeminal ganglion and thus can be considered trigeminal positional identity markers. Interestingly, establishing some of the spatial patterns requires signals from the developing face. We identified bone morphogenetic protein 4 (BMP4) as one of these target-derived factors and showed that spatially defined retrograde BMP signaling controls the differential gene expressions in trigeminal neurons through both Smad4-independent and Smad4-dependent pathways. Mice lacking one of the BMP4-regulated transcription factors, Onecut2 (OC2), have defects in the trigeminal central projections representing the whiskers. Our results provide molecular evidence for both spatial patterning and retrograde regulation of gene expression in sensory neurons during the development of the somatosensory map.
Collapse
Affiliation(s)
- Liberty K Hodge
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Xu P, Hall AK. Activin acts with nerve growth factor to regulate calcitonin gene-related peptide mRNA in sensory neurons. Neuroscience 2007; 150:665-74. [PMID: 17964731 DOI: 10.1016/j.neuroscience.2007.09.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Revised: 09/10/2007] [Accepted: 09/20/2007] [Indexed: 10/22/2022]
Abstract
Calcitonin gene-related peptide (CGRP) increases in sensory neurons after inflammation and plays an important role in abnormal pain responses, but how this neuropeptide is regulated is not well understood. Both activin A and nerve growth factor (NGF) increase in skin after inflammation and induce CGRP in neurons in vivo and in vitro. This study was designed to understand how neurons integrate these two signals to regulate the neuropeptide important for inflammatory pain. In adult dorsal root ganglion neurons, NGF but not activin alone produced a dose-dependent increase in CGRP mRNA. When added together with NGF, activin synergistically increased CGRP mRNA, indicating that sensory neurons combine these signals. Studies were then designed to learn if that combination occurred at a common receptor or shared intracellular signals. Studies with activin IB receptor or tyrosine receptor kinase A inhibitors suggested that each ligand required its cognate receptor to stimulate the neuropeptide. Further, activin did not augment NGF-initiated intracellular mitogen-activated protein kinase signals but instead stimulated Smad phosphorylation, suggesting these ligands initiated parallel signals in the cytoplasm. Activin synergy required several NGF intracellular signals to be present. Because activin did not further stimulate, but did require NGF intracellular signals, it appears that activin and NGF converge not in receptor or cytoplasmic signals, but in transcriptional mechanisms to regulate CGRP in rat sensory neurons after inflammation.
Collapse
Affiliation(s)
- P Xu
- Department of Neurosciences, Case Western Reserve University, School of Medicine,Cleveland, OH 44106, USA
| | | |
Collapse
|