1
|
Liu Y, Jin F, Zhou L, Li X, Li X, Chen Q, Yang S, Sun J, Qi F. Platelet-derived Growth Factor Receptor-α Induces Contraction Knots and Inflammatory Pain-like Behavior in a Rat Model of Myofascial Trigger Points. Anesthesiology 2024; 141:929-945. [PMID: 39058323 PMCID: PMC11463032 DOI: 10.1097/aln.0000000000005167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Myofascial trigger points (MTrPs) are the primary etiological characteristics of chronic myofascial pain syndrome. Receptor tyrosine kinases (RTKs) are associated with signal transduction in the central mechanisms of chronic pain, but the role of RTKs in the peripheral mechanisms of MTrPs remains unclear. The current study aimed to identify RTKs expression in MTrPs and elucidate the molecular mechanisms through which platelet-derived growth factor receptor-α (PDGFR-α) induces contraction knots and inflammatory pain-like behavior in a rat model of myofascial trigger points. METHODS MTrPs tissue samples were obtained from the trapezius muscles of patients with myofascial pain syndrome through needle biopsy, and PDGFR-α activation was analyzed by microarray, enzyme-linked immunosorbent assay, and histological staining. Sprague-Dawley rats (male and female) were used to investigate PDGFR-α signaling, assessing pain-like behaviors with Randall-Selitto and nest-building tests. Muscle fiber and sarcomere morphologies were observed using histology and electron microscopy. The PDGFR-α binding protein was identified by coimmunoprecipitation, liquid chromatograph mass spectrometer, and molecular docking. PDGFR-α-related protein or gene levels, muscle contraction, and inflammatory markers were determined by Western blot and reverse-transcription quantitative polymerase chain reaction. RESULTS PDGFR-α phosphorylation levels were elevated in the MTrPs tissues of individuals with trapezius muscle pain and were positively correlated with pain intensity. In rats, PDGFR-α activation caused pain-like behaviors and muscle contraction via the Janus kinase 2/signal transducer and activator of transcription-3 (JAK2/STAT3) pathway. JAK2/STAT3 inhibitors reversed the pain-like behaviors and muscle contraction induced by PDGFR-α activation. Collagen type I α 1 (COL1A1) binds to PDGFR-α and promotes its phosphorylation, which contributed to pain-like behaviors and muscle contraction. CONCLUSIONS COL1A1-induced phosphorylation of PDGFR-α and the subsequent activation of the JAK2/STAT3 pathway may induce dysfunctional muscle contraction and increased nociception at MTrPs. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Yu Liu
- Department of Anesthesiology, and Research Center for Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feihong Jin
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lingwei Zhou
- Department of Anesthesiology, and Research Center for Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuan Li
- Department of Anesthesiology, and Research Center for Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyue Li
- Department of Anesthesiology, and Research Center for Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qinghe Chen
- Department of Anesthesiology, and Research Center for Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shaozhong Yang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jintang Sun
- Research Center for Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, China
| | - Feng Qi
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
2
|
Emanuilov AI, Budnik AF, Masliukov PM. Somatostatin-immunoreactive neurons of the rat gut during the development. Histochem Cell Biol 2024; 162:385-402. [PMID: 39153131 DOI: 10.1007/s00418-024-02322-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2024] [Indexed: 08/19/2024]
Abstract
Somatostatin (SST) is a peptide expressed in the peripheral and central nervous systems, as well as in endocrine and immune cells. The aim of the current study is to determine the percentage of SST immunoreactive (IR) neurons and their colocalization with choline acetyltransferase (ChAT), neuronal nitric oxide synthase (nNOS), neuropeptide Y (NPY), and glial fibrillary acidic protein (GFAP) in the myenteric plexus (MP) and submucous plexus (SP) of the small intestine (SI) and large intestine (LI) of rats across different age groups from newborn to senescence using immunohistochemistry. In the MP of the SI and LI, the percentage of SST-IR neurons significantly increased during early postnatal development from 12 ± 2.4 (SI) and 13 ± 3.0 (LI) in newborn rats to 23 ± 1.5 (SI) and 18 ± 1.6 (LI) in 20-day-old animals, remaining stable until 60 days of age. The proportion of SST-IR cells then decreased in aged 2-year-old animals to 14 ± 2.0 (SI) and 10 ± 2.6 (LI). In the SP, the percentage of SST-IR neurons significantly rose from 22 ± 3.2 (SI) and 23 ± 1.7 (LI) in newborn rats to 42 ± 4.0 in 20-day-old animals (SI) and 32 ± 4.9 in 30-day-old animals (LI), before declining in aged 2-year-old animals to 21 ± 2.6 (SI) and 28 ± 7.4 (LI). Between birth and 60 days of age, 97-98% of SST-IR neurons in the MP and SP colocalized with ChAT in both plexuses of the SI and LI. The percentage of SST/ChAT neurons decreased in old rats to 85 ± 5.0 (SI) and 90 ± 3.8 (LI) in the MP and 89 ± 3.2 (SI) and 89 ± 1.6 (LI) in the SP. Conversely, in young rats, only a few SST-IR neurons colocalized with nNOS, but this percentage significantly increased in 2-year-old rats. The percentage of SST/NPY-IR neurons exhibited considerable variation throughout postnatal development, with no significant differences across different age groups in both the MP and SP of both intestines. No colocalization of SST with GFAP was observed in any of the studied animals. In conclusion, the expression of SST in enteric neurons increases in young rats and decreases in senescence, accompanied by changes in SST colocalization with ChAT and nNOS.
Collapse
Affiliation(s)
- Andrey I Emanuilov
- Department of Human Anatomy, Yaroslavl State Medical University, Yaroslavl, Russia
| | - Antonina F Budnik
- Department of Normal and Pathological Anatomy, Kabardino-Balkarian State University Named After H.M. Berbekov, Nalchik, Russia
| | - Petr M Masliukov
- Department of Human Anatomy, Yaroslavl State Medical University, Yaroslavl, Russia.
- Department of Normal Physiology and Biophysics, Yaroslavl State Medical University, Revoliucionnaya 5, Yaroslavl, Russia, 150000.
| |
Collapse
|
3
|
Liu X, Xu Y, Wang Y, Peng X, Jian J, Wang X, Wang T. Administration of methylprednisolone do not affect the spinal scar component of spinal cord injury. J Spinal Cord Med 2024:1-10. [PMID: 39167368 DOI: 10.1080/10790268.2024.2352929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/23/2024] Open
Abstract
OBJECTIVE The aim of this study is to evaluate the impact of methylprednisolone (MP) on scar composition following spinal cord injury (SCI). DESIGN A total of 40 adult Sprague Dawley rats underwent right hemisection injuries to the spinal cord. INTERVENTIONS The rats were randomly divided into two groups: the vehicle group and the MP group. In the MP group, rats received intraperitoneal injections of MP at a dose of 30 mg/kg for 7 consecutive days, while the vehicle group received intraperitoneal injections of saline as a control. Weekly assessments of hindlimb performance in the rat models were conducted using the Basso-Beattie-Bresnahan test (BBB) score and the horizontal ladder-walking test. Changes in scar components were identified through immunofluorescence staining, and an axonal regeneration assay was employed to evaluate regrowth under inhibitory conditions. RESULTS The administration of MP led to a significant improvement in BBB scores compared to the control group at 7 days post-injury, although this improvement was not consistent. Furthermore, rats in the MP group did not demonstrate progressive improvement in horizontal ladder walking. Notably, there were no significant changes in the content of scar components in the injured area following MP treatment, and the axon length of neurons treated with MP did not exhibit significant extension compared to the vehicle group. CONCLUSIONS Our findings indicate that the administration of MP does not effectively enhance hindlimb motor function or promote neuronal axon growth within a scarred environment after SCI.
Collapse
Affiliation(s)
- Xin Liu
- Institute of Base Medicine, Jinzhou Medical University, Jinzhou, People's Republic of China
| | - Yang Xu
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
| | - Yangyang Wang
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
| | - Xia Peng
- Institute of Neuroscience, Laboratory Zoology Department, Kunming Medical University, Kunming, People's Republic of China
| | - Jiao Jian
- Institute of Neuroscience, Laboratory Zoology Department, Kunming Medical University, Kunming, People's Republic of China
| | - Xuefang Wang
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
| | - Tinghua Wang
- Institute of Base Medicine, Jinzhou Medical University, Jinzhou, People's Republic of China
- Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu, People's Republic of China
- Institute of Neuroscience, Laboratory Zoology Department, Kunming Medical University, Kunming, People's Republic of China
| |
Collapse
|
4
|
Xiu-Ying H, Yue-Xiang Z, Hui-Si Y, Hong-Zhou Y, Qing-Jie X, Ting-Hua W. PDGFBB facilitates tumorigenesis and malignancy of lung adenocarcinoma associated with PI3K-AKT/MAPK signaling. Sci Rep 2024; 14:4191. [PMID: 38378786 PMCID: PMC10879171 DOI: 10.1038/s41598-024-54801-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
Lung adenocarcinoma (LUAD) remains one of the most aggressive tumors and the efficacy of conventional treatment has been bleak. Nowadays, gene-targeted therapy has become a new favorite in tumor therapy. Herein, we investigated the effect of platelet derived growth factor BB (PDGFBB) on LUAD. Firstly, PDGFBB was upregulated in LUAD patients and closely linked with poor survival. Furthermore, the expression of PDGFBB and PDGFRα/β in LUAD cells was higher than that in normal lung cells. By loss-of-function with herpes simplex virus (HSV)-PDGFi-shRNA, we found that PDGFBB knockdown caused a significant decrease in proliferation and migration, but evoked apoptosis of LUAD cells in vitro. Conversely, exogenous PDGFBB held adverse effect. Additionally, A549 cells with PDGFBB knockdown had a low probability of tumorigenesis in vivo. Moreover, PDGFBB knockdown restrained the growth of xenografts derived from normal A549 cells. Mechanistically, PDGFBB knockdown suppressed PI3K/AKT and Ras/MAPK signaling, while PDGFBB was the opposite. Therefore, we concluded that PDGFBB might facilitate the tumorigenesis and malignancy of LUAD through its functional downstream nodes-PI3K/AKT and Ras/MAPK signaling, which supported that PDGFBB could serve as a rational therapeutic target for LUAD.
Collapse
Affiliation(s)
- He Xiu-Ying
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zheng Yue-Xiang
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Yang Hui-Si
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Yu Hong-Zhou
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xia Qing-Jie
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Wang Ting-Hua
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China.
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China.
| |
Collapse
|
5
|
Alsaadi H, Peller J, Ghasemlou N, Kawaja MD. Immunohistochemical phenotype of sensory neurons associated with sympathetic plexuses in the trigeminal ganglia of adult nerve growth factor transgenic mice. J Comp Neurol 2024; 532:e25563. [PMID: 37986234 DOI: 10.1002/cne.25563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Following peripheral nerve injury, postganglionic sympathetic axons sprout into the affected sensory ganglia and form perineuronal sympathetic plexuses with somata of sensory neurons. This sympathosensory coupling contributes to the onset and persistence of injury-induced chronic pain. We have documented the presence of similar sympathetic plexuses in the trigeminal ganglia of adult mice that ectopically overexpress nerve growth factor (NGF), in the absence of nerve injury. In this study, we sought to further define the phenotype(s) of these trigeminal sensory neurons having sympathetic plexuses in our transgenic mice. Using quantitative immunofluorescence staining analyses, we show that the invading sympathetic axons specifically target sensory somata immunopositive for several biomarkers: NGF high-affinity receptor tyrosine kinase A (trkA), calcitonin gene-related peptide (CGRP), neurofilament heavy chain (NFH), and P2X purinoceptor 3 (P2X3). Based on these phenotypic characteristics, the majority of the sensory somata surrounded by sympathetic plexuses are likely to be NGF-responsive nociceptors (i.e., trkA expressing) that are peptidergic (i.e., CGRP expressing), myelinated (i.e., NFH expressing), and ATP sensitive (i.e., P2X3 expressing). Our data also show that very few sympathetic plexuses surround sensory somata expressing other nociceptive (pain) biomarkers, including substance P and acid-sensing ion channel 3. No sympathetic plexuses are associated with sensory somata that display isolectin B4 binding. Though the cellular mechanisms that trigger the formation of sympathetic plexus (with and without nerve injury) remain unknown, our new observations yield an unexpected specificity with which invading sympathetic axons appear to target a precise subtype of nociceptors. This selectivity likely contributes to pain development and maintenance associated with sympathosensory coupling.
Collapse
Affiliation(s)
- Hanin Alsaadi
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Jacob Peller
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Nader Ghasemlou
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, Queen's University, Kingston, Ontario, Canada
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Michael D Kawaja
- Center for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
6
|
Puig S, Gutstein HB. Chronic Morphine Modulates PDGFR-β and PDGF-B Expression and Distribution in Dorsal Root Ganglia and Spinal Cord in Male Rats. Neuroscience 2023; 519:147-161. [PMID: 36997020 DOI: 10.1016/j.neuroscience.2023.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023]
Abstract
The analgesic effect of opioids decreases over time due to the development of analgesic tolerance. We have shown that inhibition of the platelet-derived growth factor beta (PDGFR-β) signaling eliminates morphine analgesic tolerance in rats. Although the PDGFR-β and its ligand, the platelet-derived growth factor type B (PDGF-B), are expressed in the substantia gelatinosa of the spinal cord (SG) and in the dorsal root ganglia (DRG), their precise distribution within different cell types of these structures is unknown. Additionally, the impact of a tolerance-mediating chronic morphine treatment, on the expression and distribution of PDGF-B and PDGFR-β has not yet been studied. Using immunohistochemistry (IHC), we found that in the spinal cord, PDGFR-β and PDGF-B were expressed in neurons and oligodendrocytes and co-localized with the mu-opioid receptor (MOPr) in opioid naïve rats. PDGF-B was also found in microglia and astrocytes. Both PDGFR-β and PDGF-B were detected in DRG neurons but not in spinal primary afferent terminals. Chronic morphine exposure did not change the cellular distribution of PDGFR-β or PDGF-B. However, PDGFR-β expression was downregulated in the SG and upregulated in the DRG. Consistent with our previous finding that morphine caused tolerance by inducing PDGF-B release, PDGF-B was upregulated in the spinal cord. We also found that chronic morphine exposure caused a spinal proliferation of oligodendrocytes. The changes in PDGFR-β and PDGF-B expression induced by chronic morphine treatment suggest potential mechanistic substrates underlying opioid tolerance.
Collapse
Affiliation(s)
- Stephanie Puig
- Department of Pharmacology and Physiology, Boston University School of Medicine, Boston, 02118 MA, USA
| | - Howard B Gutstein
- Department of Anesthesiology, University of Connecticut Health Science Center, Farmington, 06030 CT, USA.
| |
Collapse
|
7
|
Wu P, Wang Y, Liu Y, Liu Y, Zhou G, Wu X, Wen Q. Emerging roles of the P2X7 receptor in cancer pain. Purinergic Signal 2022:10.1007/s11302-022-09902-1. [DOI: 10.1007/s11302-022-09902-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/03/2022] [Indexed: 11/21/2022] Open
Abstract
AbstractCancer pain is the most prevalent symptom experienced by cancer patients. It substantially impacts a patient’s long-term physical and emotional health, making it a pressing issue that must be addressed. Purinergic receptor P2X7 (P2X7R) is a widely distributed and potent non-selective ATP-gated ion channel that regulates tumor proliferation, chronic pain, and the formation of inflammatory lesions in the central nervous system. P2X7R plays an essential role in cancer pain and complications related to cancer pain including depression and opioid tolerance. This review focuses on the structure and distribution of P2X7R, its role in diverse tissues in cancer pain, and the application of P2X7R antagonists in the treatment of cancer pain to propose new ideas for cancer pain management.
Collapse
|
8
|
Chan WH, Huang NC, Lin YW, Lin FY, Tsai CS, Yeh CC. Intrathecal IGF2 siRNA injection provides long-lasting anti-allodynic effect in a spared nerve injury rat model of neuropathic pain. PLoS One 2021; 16:e0260887. [PMID: 34855889 PMCID: PMC8638935 DOI: 10.1371/journal.pone.0260887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 11/18/2021] [Indexed: 01/12/2023] Open
Abstract
Previous studies have shown an increase of insulin-like growth factor-2 (IGF2) in animal models of neuropathic pain. We aimed to examine the hypothesis that reducing the expression of IGF2 using intrathecal IGF2 small-interfering RNA (siRNA) would attenuate the development of neuropathic pain in rats after spared nerve injury (SNI). Male Wistar rats were divided into three groups: sham-operated group, in which surgery was performed to cut the muscles without injuring the nerves; SNI group, in which SNI surgery was performed to sever the nerves; and SNI + siRNA IGF2 group, in which SNI surgery was performed, and IGF2-siRNA was administered intrathecally 1 day after SNI. The rats were assessed for mechanical allodynia and cold allodynia 1 day before surgery (baseline), and at 2, 4, 6, 8, and 10 days after siRNA treatment. The rat spinal cord was collected for quantitative polymerase chain reaction and western blot analysis. Compared with the SNI group, rats that received IGF2 siRNA showed a significantly increased SNI-induced paw-withdrawal threshold to metal filament stimulation from Day 4 to Day 10 after SNI surgery. IGF2 siRNA significantly decreased the response duration from the acetone test from Day 2 to Day 10 following SNI surgery. SNI increased IGF2 mRNA expression on Day 2 and increased IGF2 protein expression on Day 8 and Day 10 in the spinal cord of the SNI rats. However, the above-mentioned effects of IGF2 mRNA and protein expression were significantly inhibited in the SNI + IGF2 siRNA group. We demonstrated that intrathecal administration of IGF2 siRNA provided significant inhibition of SNI-induced neuropathic pain via inhibition of IGF2 expression in the spinal cord. The analgesic effect lasted for 10 days. Further exploration of intrathecal IGF2 siRNA administration as a potential therapeutic strategy for neuropathic pain is warranted.
Collapse
Affiliation(s)
- Wei-Hung Chan
- Department of Anesthesiology, National Defense Medical Center, Taipei, Taiwan
- Department of Anesthesiology, Tri-Service General Hospital, Taipei, Taiwan
| | - Nian-Cih Huang
- Department of Anesthesiology, National Defense Medical Center, Taipei, Taiwan
- Department of Anesthesiology, Tri-Service General Hospital, Taipei, Taiwan
| | - Yi-Wen Lin
- Institute of Oral Biology, National Yang-Ming Chiao-Tung University, Hsinchu, Taiwan
| | - Feng-Yen Lin
- Department of Internal Medicine and Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Chang Yeh
- Department of Anesthesiology, National Defense Medical Center, Taipei, Taiwan
- Department of Anesthesiology, Tri-Service General Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
9
|
Gadepalli A, Akhilesh, Uniyal A, Modi A, Chouhan D, Ummadisetty O, Khanna S, Solanki S, Allani M, Tiwari V. Multifarious Targets and Recent Developments in the Therapeutics for the Management of Bone Cancer Pain. ACS Chem Neurosci 2021; 12:4195-4208. [PMID: 34723483 DOI: 10.1021/acschemneuro.1c00414] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Bone cancer pain (BCP) is a distinct pain state showing characteristics of both neuropathic and inflammatory pain. On average, almost 46% of cancer patients exhibit BCP with numbers flaring up to as high as 76% for terminally ill patients. Patients suffering from BCP experience a compromised quality of life, and the unavailability of effective therapeutics makes this a more devastating condition. In every individual cancer patient, the pain is driven by different mechanisms at different sites. The mechanisms behind the manifestation of BCP are very complex and poorly understood, which creates a substantial barrier to drug development. Nevertheless, some of the key mechanisms involved have been identified and are being explored further to develop targeted molecules. Developing a multitarget approach might be beneficial in this case as the underlying mechanism is not fixed and usually a number of these pathways are simultaneously dysregulated. In this review, we have discussed the role of recently identified novel modulators and mechanisms involved in the development of BCP. They include ion channels and receptors involved in sensing alteration of temperature and acidic microenvironment, immune system activation, sodium channels, endothelins, protease-activated receptors, neurotrophins, motor proteins mediated trafficking of glutamate receptor, and some bone-specific mechanisms. Apart from this, we have also discussed some of the novel approaches under preclinical and clinical development for the treatment of bone cancer pain.
Collapse
Affiliation(s)
- Anagha Gadepalli
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Ankit Uniyal
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Ajay Modi
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Obulapathi Ummadisetty
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Shreya Khanna
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Shreya Solanki
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Meghana Allani
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| |
Collapse
|
10
|
Borges JP, Mekhail K, Fairn GD, Antonescu CN, Steinberg BE. Modulation of Pathological Pain by Epidermal Growth Factor Receptor. Front Pharmacol 2021; 12:642820. [PMID: 34054523 PMCID: PMC8149758 DOI: 10.3389/fphar.2021.642820] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic pain has been widely recognized as a major public health problem that impacts multiple aspects of patient quality of life. Unfortunately, chronic pain is often resistant to conventional analgesics, which are further limited by their various side effects. New therapeutic strategies and targets are needed to better serve the millions of people suffering from this devastating disease. To this end, recent clinical and preclinical studies have implicated the epidermal growth factor receptor signaling pathway in chronic pain states. EGFR is one of four members of the ErbB family of receptor tyrosine kinases that have key roles in development and the progression of many cancers. EGFR functions by activating many intracellular signaling pathways following binding of various ligands to the receptor. Several of these signaling pathways, such as phosphatidylinositol 3-kinase, are known mediators of pain. EGFR inhibitors are known for their use as cancer therapeutics but given recent evidence in pilot clinical and preclinical investigations, may have clinical use for treating chronic pain. Here, we review the clinical and preclinical evidence implicating EGFR in pathological pain states and provide an overview of EGFR signaling highlighting how EGFR and its ligands drive pain hypersensitivity and interact with important pain pathways such as the opioid system.
Collapse
Affiliation(s)
- Jazlyn P Borges
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Katrina Mekhail
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Gregory D Fairn
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Costin N Antonescu
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Benjamin E Steinberg
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
11
|
Li Z, Peng X, Jia X, Su P, Liu D, Tu Y, Xu Q, Gao F. Spinal heat shock protein 27 participates in PDGFRβ-mediated morphine tolerance through PI3K/Akt and p38 MAPK signalling pathways. Br J Pharmacol 2020; 177:5046-5062. [PMID: 32559815 PMCID: PMC7589020 DOI: 10.1111/bph.15169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The development of antinociceptive morphine tolerance is a clinically intractable problem. Earlier work has demonstrated the pivotal roles of PDGF and its receptor PDGFRβ in morphine tolerance. Here, we have investigated the role of spinal heat shock protein 27 (HSP27) in morphine tolerance and its relationship with PDGFRβ activation. EXPERIMENTAL APPROACH Rats were treated with morphine for 9 days, and its anti-nociceptive effect against thermal pain was evaluated by a tail-flick latency test. Western blot, real-time PCR, immunofluorescent staining, and various antagonists, agonists, and siRNA lentiviral vectors elucidated the roles of HSP27, PDGFRβ, and related signalling pathways in morphine tolerance. KEY RESULTS Chronic morphine administration increased expression and phosphorylation of HSP27 in the spinal cord. Down-regulating HSP27 attenuated the development of morphine tolerance. PDGFRβ antagonism inhibited HSP27 activation and attenuated and reversed morphine tolerance. PDGFRβ induction increased HSP27 expression and activation and partly decreased morphine analgesia. PDGFRβ inhibition reduced Akt and p38 MAPK activity in morphine tolerance. PI3K and p38 inhibitors reversed morphine tolerance and suppressed morphine-induced HSP27 phosphorylation. CONCLUSION AND IMPLICATIONS This study demonstrated for the first time that spinal HSP27 participates in PDGFRβ-mediated morphine tolerance via the PI3K/Akt and p38 MAPK signalling pathways. These findings suggest a potential clinical strategy for prolonging the antinociceptive effects of opioids during long-term pain control.
Collapse
Affiliation(s)
- Zheng Li
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoling Peng
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoqian Jia
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Peng Su
- Department of Anesthesiology, Sichuan Academy of Medical SciencesSichuan Provincial People's HospitalChengduChina
| | - Daiqiang Liu
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Ye Tu
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Qiaoqiao Xu
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Feng Gao
- Department of Anesthesiology, Tongji HospitalHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
12
|
Li Z, Jia X, Peng X, Gao F. The Interaction Between Spinal PDGFRβ and μ Opioid Receptor in the Activation of Microglia in Morphine-Tolerant Rats. J Pain Res 2020; 13:1803-1810. [PMID: 32765055 PMCID: PMC7381827 DOI: 10.2147/jpr.s255221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose Opioid tolerance remains a challenging problem, which limits prolonged drug usage in clinics. Previous studies have shown a fundamental role of platelet-derived growth factor receptor β submit (PDGFRβ) in morphine tolerance. The aim of this study was to investigate the mechanisms of spinal PDGFRβ activation in morphine tolerance. Methods Rats were treated with morphine for 7 days and the effect of drug was evaluated by tail-flick latency test. By using Western blot and real-time PCR, the interaction between μ opioid receptor (MOR) and PDGFRβ in microglia activation, as well as related signaling pathways during morphine tolerance were investigated. Results Chronic PDGFRβ agonist could induce microglia activation in spinal cord and decrease the analgesic effect of morphine. PDGFRβ inhibitor suppressed microglia activation during the development of morphine tolerance. Furthermore, antagonizing MOR could effectively inhibit the phosphorylations of PDGFRβ and JNK. Blocking PDGFRβ had no influence on JNK signaling, while JNK inhibitor could decrease the phosphorylation of PDGFRβ. Conclusion These results provide direct evidence that repeatedly activating MOR by morphine could induce the transactivation of PDGFRβ via JNK MAPK in spinal cord, which leads to microglia activation during the development of morphine tolerance.
Collapse
Affiliation(s)
- Zheng Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiaoqian Jia
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiaoling Peng
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
13
|
Ma S, Zheng X, Zheng T, Huang F, Jiang J, Luo H, Guo Q, Hu B. Amitriptyline influences the mechanical withdrawal threshold in bone cancer pain rats by regulating glutamate transporter GLAST. Mol Pain 2020; 15:1744806919855834. [PMID: 31218920 PMCID: PMC6637840 DOI: 10.1177/1744806919855834] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Patients with cancer, especially breast, prostate, and lung cancer, commonly experience bone metastases that are difficult to manage and are associated with bone cancer pain. Amitriptyline is often used to treat chronic pain, such as neuropathic pain. In this study, the effects of amitriptyline on the mechanical withdrawal threshold and its underlying mechanisms were evaluated in rat models of bone cancer pain. Walker 256 rat mammary gland carcinoma cells were injected into the bone marrow cavity of the right tibia of rats to provoke bone cancer pain. Then, amitriptyline was intraperitoneally administered twice daily from fifth day after the operation. Rats with bone cancer showed an apparent decline in the mechanical withdrawal threshold at day 11 after Walker 256 cells inoculation. The levels of the glutamate-aspartate transporter in the spinal cord dorsal horn decreased remarkably, and the concentration of the excitatory amino acid glutamate in the cerebrospinal fluid increased substantially. Amitriptyline injection could prevent the decline of mechanical withdrawal threshold in bone cancer pain rats. In addition, glutamate-aspartate transporter was upregulated on the glial cell surface, and glutamate levels were reduced in the cerebrospinal fluid. However, amitriptyline injection could not prevent the bone cancer pain-induced reduction in glutamate-aspartate transporter in the glial cell cytosol, it further downregulated cytosolic glutamate-aspartate transporter. Amitriptyline had no significant effect on GLAST messenger RNA expression, and bone cancer pain-invoked protein kinase A/protein kinase C upregulation was prevented. Taken together, these results suggest that the intraperitoneal injection of amitriptyline can prevent the decrease of mechanical withdrawal threshold in bone cancer pain rats, the underlying mechanisms may be associated with the inhibition of protein kinase A/protein kinase C expression, thus promoting glutamate-aspartate transporter trafficking onto the glial cell surface and reducing excitatory amino acid concentrations in the cerebrospinal fluid.
Collapse
Affiliation(s)
- Simeng Ma
- 1 Fujian Provincial Hospital, Fuzhou, China
| | | | - Ting Zheng
- 1 Fujian Provincial Hospital, Fuzhou, China
| | | | | | | | | | - Bin Hu
- 1 Fujian Provincial Hospital, Fuzhou, China
| |
Collapse
|
14
|
Platelet-derived growth factor activates nociceptive neurons by inhibiting M-current and contributes to inflammatory pain. Pain 2020; 160:1281-1296. [PMID: 30933959 PMCID: PMC6553959 DOI: 10.1097/j.pain.0000000000001523] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Supplemental Digital Content is Available in the Text. Our work reveals that the platelet-derived growth factor-BB, by inhibiting nociceptive M-type potassium channels, acts as a pain-inducing proinflammatory factor that significantly contributes to inflammatory pain. Endogenous inflammatory mediators contribute to the pathogenesis of pain by acting on nociceptors, specialized sensory neurons that detect noxious stimuli. Here, we describe a new factor mediating inflammatory pain. We show that platelet-derived growth factor (PDGF)-BB applied in vitro causes repetitive firing of dissociated nociceptor-like rat dorsal root ganglion neurons and decreased their threshold for action potential generation. Injection of PDGF-BB into the paw produced nocifensive behavior in rats and led to thermal and mechanical pain hypersensitivity. We further detailed the biophysical mechanisms of these PDGF-BB effects and show that PDGF receptor–induced inhibition of nociceptive M-current underlies PDGF-BB–mediated nociceptive hyperexcitability. Moreover, in vivo sequestration of PDGF or inhibition of the PDGF receptor attenuates acute formalin-induced inflammatory pain. Our discovery of a new pain-facilitating proinflammatory mediator, which by inhibiting M-current activates nociceptive neurons and thus contributes to inflammatory pain, improves our understanding of inflammatory pain pathophysiology and may have important clinical implications for pain treatment.
Collapse
|
15
|
de Almeida AS, Rigo FK, De Prá SDT, Milioli AM, Dalenogare DP, Pereira GC, Ritter CDS, Peres DS, Antoniazzi CTDD, Stein C, Moresco RN, Oliveira SM, Trevisan G. Characterization of Cancer-Induced Nociception in a Murine Model of Breast Carcinoma. Cell Mol Neurobiol 2019; 39:605-617. [PMID: 30850915 DOI: 10.1007/s10571-019-00666-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 02/25/2019] [Indexed: 12/14/2022]
Abstract
Severe and poorly treated pain often accompanies breast cancer. Thus, novel mechanisms involved in breast cancer-induced pain should be investigated. Then, it is necessary to characterize animal models that are reliable with the symptoms and progression of the disease as observed in humans. Explaining cancer-induced nociception in a murine model of breast carcinoma was the aim of this study. 4T1 (104) lineage cells were inoculated in the right fourth mammary fat pad of female BALB/c mice; after this, mechanical and cold allodynia, or mouse grimace scale (MGS) were observed for 30 days. To determine the presence of bone metastasis, we performed the metastatic clonogenic test and measure calcium serum levels. At 20 days after tumor induction, the antinociceptive effect of analgesics used to relieve pain in cancer patients (acetaminophen, naproxen, codeine or morphine) or a cannabinoid agonist (WIN 55,212-2) was tested. Mice inoculated with 4T1 cells developed mechanical and cold allodynia and increased MGS. Bone metastasis was confirmed using the clonogenic assay, and hypercalcemia was observed 20 days after cells inoculation. All analgesic drugs reduced the mechanical and cold allodynia, while the MGS was decreased only by the administration of naproxen, codeine, or morphine. Also, WIN 55,212-2 improved all nociceptive measures. This pain model could be a reliable form to observe the mechanisms of breast cancer-induced pain or to observe the efficacy of novel analgesic compounds.
Collapse
Affiliation(s)
- Amanda Spring de Almeida
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Flávia Karine Rigo
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), Criciúma, SC, 88006-000, Brazil
| | - Samira Dal-Toé De Prá
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), Criciúma, SC, 88006-000, Brazil
| | - Alessandra Marcone Milioli
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), Criciúma, SC, 88006-000, Brazil
| | - Diéssica Padilha Dalenogare
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Gabriele Cheiran Pereira
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Camila Dos Santos Ritter
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Diulle Spat Peres
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | | | - Carolina Stein
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Rafael Noal Moresco
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Sara Marchesan Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Gabriela Trevisan
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil.
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), Criciúma, SC, 88006-000, Brazil.
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Avenida Roraima, 1000, Building 21, Room 5207, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
16
|
de la Peña JBI, Song JJ, Campbell ZT. RNA control in pain: Blame it on the messenger. WILEY INTERDISCIPLINARY REVIEWS-RNA 2019; 10:e1546. [PMID: 31090211 DOI: 10.1002/wrna.1546] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/17/2019] [Accepted: 04/22/2019] [Indexed: 12/12/2022]
Abstract
mRNA function is meticulously controlled. We provide an overview of the integral role that posttranscriptional controls play in the perception of painful stimuli by sensory neurons. These specialized cells, termed nociceptors, precisely regulate mRNA polarity, translation, and stability. A growing body of evidence has revealed that targeted disruption of mRNAs and RNA-binding proteins robustly diminishes pain-associated behaviors. We propose that the use of multiple independent regulatory paradigms facilitates robust temporal and spatial precision of protein expression in response to a range of pain-promoting stimuli. This article is categorized under: RNA in Disease and Development > RNA in Disease Translation > Translation Regulation RNA Turnover and Surveillance > Regulation of RNA Stability.
Collapse
Affiliation(s)
- June Bryan I de la Peña
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| | - Jane J Song
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| | - Zachary T Campbell
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| |
Collapse
|
17
|
Diaz-delCastillo M, Christiansen SH, Appel CK, Falk S, Woldbye DP, Heegaard AM. Neuropeptide Y is Up-regulated and Induces Antinociception in Cancer-induced Bone Pain. Neuroscience 2018; 384:111-119. [DOI: 10.1016/j.neuroscience.2018.05.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 04/18/2018] [Accepted: 05/18/2018] [Indexed: 01/29/2023]
|
18
|
Chen NF, Sung CS, Wen ZH, Chen CH, Feng CW, Hung HC, Yang SN, Tsui KH, Chen WF. Therapeutic Effect of Platelet-Rich Plasma in Rat Spinal Cord Injuries. Front Neurosci 2018; 12:252. [PMID: 29740270 PMCID: PMC5924817 DOI: 10.3389/fnins.2018.00252] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/03/2018] [Indexed: 11/13/2022] Open
Abstract
Platelet-rich plasma (PRP) is prepared by centrifuging fresh blood in an anticoagulant state, and harvesting the platelet-rich portion or condensing platelets. Studies have consistently demonstrated that PRP concentrates are an abundant source of growth factors, such as platelet-derived growth factor (PDGF), transforming growth factor β (TGF-β), insulin-like growth factor 1 (IGF-1), and epithelial growth factor (EGF). The complex mechanisms underlying spinal cord injury (SCI) diminish intrinsic repair and neuronal regeneration. Several studies have suggested that growth factor-promoted axonal regeneration can occur for an extended period after injury. More importantly, the delivery of exogenous growth factors contained in PRP, such as EGF, IGF-1, and TGF-β, has neurotrophic effects on central nervous system (CNS) injuries and neurodegenerative diseases. However, only a few studies have investigated the effects of PRP on CNS injuries or neurodegenerative diseases. According to our review of relevant literature, no study has investigated the effect of intrathecal (i.t.) PRP injection into the injured spinal cord and activation of intrinsic mechanisms. In the present study, we directly injected i.t. PRP into rat spinal cords and examined the effects of PRP on normal and injured spinal cords. In rats with normal spinal cords, PRP induced microglia and astrocyte activation and PDGF-B and ICAM-1 expression. In rats with SCIs, i.t. PRP enhanced the locomotor recovery and spared white matter, promoted angiogenesis and neuronal regeneration, and modulated blood vessel size. Furthermore, a sustained treatment (a bolus of PRP followed by a 1/3 dose of initial PRP concentration) exerted more favorable therapeutic effects than a single dose of PRP. Our findings suggest by i.t. PRP stimulate angiogenesis, enhancing neuronal regeneration after SCI in rats. Although PRP induces minor inflammation in normal and injured spinal cords, it has many advantages. It is an autologous, biocompatible, nontoxic material that does not result in a major immune response. In addition, based on its safety and ease of preparation, we hypothesize that PRP is a promising therapeutic agent for SCI.
Collapse
Affiliation(s)
- Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan.,Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Sung Sung
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Zhi-Hong Wen
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chun-Hong Chen
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei, Taiwan
| | - Chien-Wei Feng
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei, Taiwan
| | - Han-Chun Hung
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei, Taiwan
| | - San-Nan Yang
- School of Medicine, College of Medicine and Department of Pediatrics, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Department of Obstetrics and Gynecology and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Biological Science, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County, Taiwan
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Neurosurgery, Xiamen Chang Gung Hospital, Fujian, China
| |
Collapse
|
19
|
Yang Y, Fang S. Small non-coding RNAs-based bone regulation and targeting therapeutic strategies. Mol Cell Endocrinol 2017; 456:16-35. [PMID: 27888003 PMCID: PMC7116989 DOI: 10.1016/j.mce.2016.11.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/06/2016] [Accepted: 11/21/2016] [Indexed: 01/08/2023]
Abstract
Small non-coding RNAs, which are 20-25 nucleotide ribonucleic acids, have emerged as an important transformation in the biological evolution over almost three decades. microRNAs (miRNAs) and short interfering RNAs (siRNAs) are two significant categories of the small RNAs that exert important effects on bone endocrinology and skeletology. Therefore, clarifying the expression and function of these important molecules in bone endocrine physiology and pathology is of great significance for improving their potential therapeutic value for metabolism-associated bone diseases. In the present review, we highlight the recent advances made in understanding the function and molecular mechanism of these small non-coding RNAs in bone metabolism, especially their potentially therapeutic values in bone-related diseases.
Collapse
Affiliation(s)
- Ying Yang
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, China
| | - Sijie Fang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, China.
| |
Collapse
|