1
|
Giri SS, Tripathi AS, Erkekoğlu P, Zaki MEA. Molecular pathway of pancreatic cancer-associated neuropathic pain. J Biochem Mol Toxicol 2024; 38:e23638. [PMID: 38613466 DOI: 10.1002/jbt.23638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 04/15/2024]
Abstract
The pancreas is a heterocrine gland that has both exocrine and endocrine parts. Most pancreatic cancer begins in the cells that line the ducts of the pancreas and is called pancreatic ductal adenocarcinoma (PDAC). PDAC is the most encountered pancreatic cancer type. One of the most important characteristic features of PDAC is neuropathy which is primarily due to perineural invasion (PNI). PNI develops tumor microenvironment which includes overexpression of fibroblasts cells, macrophages, as well as angiogenesis which can be responsible for neuropathy pain. In tumor microenvironment inactive fibroblasts are converted into an active form that is cancer-associated fibroblasts (CAFs). Neurotrophins they also increase the level of Substance P, calcitonin gene-related peptide which is also involved in pain. Matrix metalloproteases are the zinc-associated proteases enzymes which activates proinflammatory interleukin-1β into its activated form and are responsible for release and activation of Substance P which is responsible for neuropathic pain by transmitting pain signal via dorsal root ganglion. All the molecules and their role in being responsible for neuropathic pain are described below.
Collapse
Affiliation(s)
| | - Alok Shiomurti Tripathi
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, Uttar Pradesh, India
| | - Pınar Erkekoğlu
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Magdi E A Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Tazawa R, Kenmoku T, Uchida K, Arendt-Nielsen L, Nagura N, Nakawaki M, Matsumoto T, Inoue G, Takeuchi H, Jimbo T, Nakazawa T, Fukuda M, Takaso M. Increased nerve growth factor expression in the synovial tissues of patients with rotator cuff tears. Mol Pain 2021; 17:17448069211021252. [PMID: 34074169 PMCID: PMC8175831 DOI: 10.1177/17448069211021252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background Rotator cuff tears (RCTs) are often associated with severe shoulder pain. Non-steroidal anti-inflammatory drugs, not recommended for long-term use, do not effectively manage RCT-induced pain, resulting in reduced quality of life. To improve management, a better understanding of the fundamental properties of RCT pain is needed. Here, we aimed to compare the expression levels of nerve growth factor (NGF) and cyclooxygenase-2 (COX-2) mRNA in the synovial tissues of patients with RCT-induced pain and patients with non-painful recurrent shoulder dislocation (RSD). Methods The study included 32 patients with RCT who underwent arthroscopic rotator cuff repair and 28 patients with non-painful RSD who underwent arthroscopic Bankart repair. Synovial tissue samples were harvested from subacromial bursa and rotator interval of RCT patients and from the rotator interval of RSD patients. Samples were analyzed quantitatively expression levels for NGF and COX2 mRNA and NGF protein. Results NGF mRNA and protein levels were significantly higher in the rotator interval of RCT patients than in the rotator interval of RSD patients (p = 0.0017, p = 0.012, respectively), while COX2 mRNA levels did not differ significantly between the two patient groups. In RCT patients, COX2 mRNA was more highly expressed in the rotator interval than in the subacromial bursa (p = 0.038), whereas the mRNA and protein levels of NGF did not differ between the two tissues. The expression of NGF mRNA in the synovium of the rotator interval was significantly correlated with the numeric rating scale of pain (ρ = 0.38, p = 0.004). Conclusion NGF mRNA and protein levels were elevated in patients with painful RCT compared with those in patients with non-painful RSD, whereas COX-2 levels were comparable in the two patient groups. These findings provide insights into novel potential strategies for clinical management of RCT.
Collapse
Affiliation(s)
- Ryo Tazawa
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Tomonori Kenmoku
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kentaro Uchida
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Lars Arendt-Nielsen
- Department of Health Science and Technology, Center for Neuroplasticity and Pain, School of Medicine, Aalborg University, Aalborg, Denmark
| | - Naoshige Nagura
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Mitsufumi Nakawaki
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Toshihide Matsumoto
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Gen Inoue
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hiroto Takeuchi
- Department of Rehabilitation, Kitasato University School of Medicine, Sagamihara, Japan
| | - Takenori Jimbo
- Department of Rehabilitation, Kitasato University School of Medicine, Sagamihara, Japan
| | - Toshiyuki Nakazawa
- Department of Rehabilitation, Kitasato University School of Medicine, Sagamihara, Japan
| | - Michinari Fukuda
- Department of Rehabilitation, Kitasato University School of Medicine, Sagamihara, Japan
| | - Masashi Takaso
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
3
|
Gu Y, Cao H, Li F, Yu J, Nian R, Feng D, Lin J, Song H, Liu W. Production of functional human nerve growth factor from the submandibular glands of mice using a CRISPR/Cas9 genome editing system. World J Microbiol Biotechnol 2020; 36:176. [PMID: 33103226 DOI: 10.1007/s11274-020-02951-x] [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: 06/15/2020] [Accepted: 10/14/2020] [Indexed: 11/24/2022]
Abstract
Nerve growth factor (NGF) is an essential trophic factor for the growth and survival of neurons in the central and peripheral nervous systems. For many years, mouse NGF (mNGF) has been used to treat various neuronal and non-neuronal disorders. However, the biological activity of human NGF (hNGF) is significantly higher than that of mNGF in human cells. Using the CRISPR/Cas9 system, we constructed the transgenic mice expressing hNGF specifically in their submandibular glands. As demonstrated by fluorescence immunohistochemical staining, these mice produced hNGF successfully, with 0.8 mg produced per gram of submandibular glands. hNGF with 99% purity was successfully extracted by two-step ion-exchange chromatography and one-step size-exclusion chromatography from the submandibular glands of these transgenic mice. Further, the purified hNGF was verified by LC-MS/MS. We analyzed the NH2-terminus of hNGF using both Edman degradation and LC-MS/MS-based methods. Both results showed that the obtained hNGF lost the NH2-terminal octapeptide (SSSHPIFH). Moreover, the produced hNGF demonstrated a strong promotion in the proliferation of TF1 cells.
Collapse
Affiliation(s)
- Yi Gu
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189 Songling Road, Qingdao, 266101, China.,University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing, 100049, China
| | - Hui Cao
- Shenzhen Innova Nanobodi Co., Ltd, No. 7018 Caitian Road, Shenzhen, 518000, China
| | - Fei Li
- Shenzhen Innova Nanobodi Co., Ltd, No. 7018 Caitian Road, Shenzhen, 518000, China
| | - Jianli Yu
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189 Songling Road, Qingdao, 266101, China.,University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing, 100049, China
| | - Rui Nian
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189 Songling Road, Qingdao, 266101, China
| | - Dongxiao Feng
- Shenzhen Innova Nanobodi Co., Ltd, No. 7018 Caitian Road, Shenzhen, 518000, China
| | - Jingtao Lin
- Dalang Hospital of Dongguan, No. 85 Jinlangzhong Road, Dalang, 523770, Dongguan, China
| | - Haipeng Song
- Shenzhen Innova Nanobodi Co., Ltd, No. 7018 Caitian Road, Shenzhen, 518000, China.
| | - Wenshuai Liu
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189 Songling Road, Qingdao, 266101, China.
| |
Collapse
|
4
|
Sung K, Ferrari LF, Yang W, Chung C, Zhao X, Gu Y, Lin S, Zhang K, Cui B, Pearn ML, Maloney MT, Mobley WC, Levine JD, Wu C. Swedish Nerve Growth Factor Mutation (NGF R100W) Defines a Role for TrkA and p75 NTR in Nociception. J Neurosci 2018; 38:3394-3413. [PMID: 29483280 PMCID: PMC5895035 DOI: 10.1523/jneurosci.1686-17.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 01/23/2018] [Accepted: 02/12/2018] [Indexed: 12/14/2022] Open
Abstract
Nerve growth factor (NGF) exerts multiple functions on target neurons throughout development. The recent discovery of a point mutation leading to a change from arginine to tryptophan at residue 100 in the mature NGFβ sequence (NGFR100W) in patients with hereditary sensory and autonomic neuropathy type V (HSAN V) made it possible to distinguish the signaling mechanisms that lead to two functionally different outcomes of NGF: trophic versus nociceptive. We performed extensive biochemical, cellular, and live-imaging experiments to examine the binding and signaling properties of NGFR100W Our results show that, similar to the wild-type NGF (wtNGF), the naturally occurring NGFR100W mutant was capable of binding to and activating the TrkA receptor and its downstream signaling pathways to support neuronal survival and differentiation. However, NGFR100W failed to bind and stimulate the 75 kDa neurotrophic factor receptor (p75NTR)-mediated signaling cascades (i.e., the RhoA-Cofilin pathway). Intraplantar injection of NGFR100W into adult rats induced neither TrkA-mediated thermal nor mechanical acute hyperalgesia, but retained the ability to induce chronic hyperalgesia based on agonism for TrkA signaling. Together, our studies provide evidence that NGFR100W retains trophic support capability through TrkA and one aspect of its nociceptive signaling, but fails to engage p75NTR signaling pathways. Our findings suggest that wtNGF acts via TrkA to regulate the delayed priming of nociceptive responses. The integration of both TrkA and p75NTR signaling thus appears to regulate neuroplastic effects of NGF in peripheral nociception.SIGNIFICANCE STATEMENT In the present study, we characterized the naturally occurring nerve growth factor NGFR100W mutant that is associated with hereditary sensory and autonomic neuropathy type V. We have demonstrated for the first time that NGFR100W retains trophic support capability through TrkA, but fails to engage p75NTR signaling pathways. Furthermore, after intraplantar injection into adult rats, NGFR100W induced neither thermal nor mechanical acute hyperalgesia, but retained the ability to induce chronic hyperalgesia. We have also provided evidence that the integration of both TrkA- and p75NTR-mediated signaling appears to regulate neuroplastic effects of NGF in peripheral nociception. Our study with NGFR100W suggests that it is possible to uncouple trophic effect from nociceptive function, both induced by wild-type NGF.
Collapse
Affiliation(s)
| | - Luiz F Ferrari
- Department of Oral Surgery, University of California San Francisco, San Francisco, California 94143
| | - Wanlin Yang
- Department of Neurosciences
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China 200025
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 143-701, South Korea
| | | | - Yingli Gu
- Department of Neurosciences
- Department of Neurology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, China 150001
| | - Suzhen Lin
- Department of Neurosciences
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China 200025
| | - Kai Zhang
- Department of Chemistry
- Department of Biochemistry, Neuroscience Program, Center for Biophysics and Quantitative Biology, Chemistry-Biology Interface Training Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, and
| | | | - Matthew L Pearn
- Department of Anesthesiology, University of California San Diego, School of Medicine, La Jolla, California 92093
- V.A. San Diego Healthcare System, San Diego, California 92161
| | - Michael T Maloney
- Department of Neurosciences, Stanford University, Stanford, California 94305
| | | | - Jon D Levine
- Department of Oral Surgery, University of California San Francisco, San Francisco, California 94143
| | - Chengbiao Wu
- Department of Neurosciences,
- V.A. San Diego Healthcare System, San Diego, California 92161
| |
Collapse
|
5
|
Malty RH, Hudmon A, Fehrenbacher JC, Vasko MR. Long-term exposure to PGE2 causes homologous desensitization of receptor-mediated activation of protein kinase A. J Neuroinflammation 2016; 13:181. [PMID: 27400965 PMCID: PMC4940832 DOI: 10.1186/s12974-016-0645-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/27/2016] [Indexed: 02/07/2023] Open
Abstract
Background Acute exposure to prostaglandin E2 (PGE2) activates EP receptors in sensory neurons which triggers the cAMP-dependent protein kinase A (PKA) signaling cascade resulting in enhanced excitability of the neurons. With long-term exposure to PGE2, however, the activation of PKA does not appear to mediate persistent PGE2-induced sensitization. Consequently, we examined whether homologous desensitization of PGE2-mediated PKA activation occurs after long-term exposure of isolated sensory neurons to the eicosanoid. Methods Sensory neuronal cultures were harvested from the dorsal root ganglia of adult male Sprague-Dawley rats. The cultures were pretreated with vehicle or PGE2 and used to examine signaling mechanisms mediating acute versus persistent sensitization by exposure to the eicosanoid using enhanced capsaicin-evoked release of immunoreactive calcitonin gene-related peptide (iCGRP) as an endpoint. Neuronal cultures chronically exposed to vehicle or PGE2 also were used to study the ability of the eicosanoid and other agonists to activate PKA and whether long-term exposure to the prostanoid alters expression of EP receptor subtypes. Results Acute exposure to 1 μM PGE2 augments the capsaicin-evoked release of iCGRP, and this effect is blocked by the PKA inhibitor H-89. After 5 days of exposure to 1 μM PGE2, administration of the eicosanoid still augments evoked release of iCGRP, but the effect is not attenuated by inhibition of PKA or by inhibition of PI3 kinases. The sensitizing actions of PGE2 after acute and long-term exposure were attenuated by EP2, EP3, and EP4 receptor antagonists, but not by an EP1 antagonist. Exposing neuronal cultures to 1 μM PGE2 for 12 h to 5 days blocks the ability of PGE2 to activate PKA. The offset of the desensitization occurs within 24 h of removal of PGE2 from the cultures. Long-term exposure to PGE2 also results in desensitization of the ability of a selective EP4 receptor agonist, L902688 to activate PKA, but does not alter the ability of cholera toxin, forskolin, or a stable analog of prostacyclin to activate PKA. Conclusions Long-term exposure to PGE2 results in homologous desensitization of EP4 receptor activation of PKA, but not to neuronal sensitization suggesting that activation of PKA does not mediate PGE2-induced sensitization after chronic exposure to the eicosanoid.
Collapse
Affiliation(s)
- Ramy Habashy Malty
- Department of Chemistry and Biochemistry, Faculty of Science, University of Regina, Regina, SK, Canada.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andy Hudmon
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jill C Fehrenbacher
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael R Vasko
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, 635 Barnhill Dr., A449, Indianapolis, IN, 46202, USA.
| |
Collapse
|
6
|
Ängeby Möller K, Berge OG, Finn A, Stenfors C, Svensson CI. Using gait analysis to assess weight bearing in rats with Freund׳s complete adjuvant-induced monoarthritis to improve predictivity: Interfering with the cyclooxygenase and nerve growth factor pathways. Eur J Pharmacol 2015; 756:75-84. [PMID: 25792342 DOI: 10.1016/j.ejphar.2015.02.050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/30/2015] [Accepted: 02/03/2015] [Indexed: 12/18/2022]
Abstract
Lack of predictive power for drug effects has been a major criticism against animal pain models. It is therefore important to define the utility and limitations of different models. The aim of this study was to extend previous work on gait analysis as a tool to investigate pharmacological effects in monoarthritic rats, specifically to test the hypothesis that monoarthritis induced by Freund׳s complete adjuvant (FCA) provides a better estimate of overall analgesic efficacy of established, and novel, clinically effective and ineffective therapeutic approaches. Male rats injected intra-articularly into one ankle joint with FCA (1.0mg/ml) were treated with the monoclonal antibody to nerve growth factor (NGF), MEDI-578, the inhibitors of tropomyosin receptor kinases A, B and C (pan-Trk) AZ6623 or AZ7092, the transient receptor potential vanilloid 1 (TRPV1) inhibitor AZD1386, or the cyclooxygenase (COX) inhibitors naproxen, ibuprofen, valdecoxib or rofecoxib. Effects on weight bearing during locomotion were tested using video capture of print images. The apparent efficacy in this model was Trk inhibitors≥anti-NGF antibody>COX inhibitors. The TRPV1 inhibitor was ineffective. Together with previous data, the results support using gait-related parameters in the monoarthritis model. FCA as induction agent seems to provide a good overall prediction of analgesic efficacy in disorders with inflammatory joint pain.
Collapse
Affiliation(s)
- Kristina Ängeby Möller
- AstraZeneca R&D Södertälje, CNSP iMed Science, SE-151 85 Södertälje, Sweden; Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| | - Odd-Geir Berge
- AstraZeneca R&D Södertälje, CNSP iMed Science, SE-151 85 Södertälje, Sweden; Multidisciplinary Pain Center, Uppsala University Hospital, SE-751 85 Uppsala, Sweden
| | - Anja Finn
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Carina Stenfors
- AstraZeneca R&D Södertälje, CNSP iMed Science, SE-151 85 Södertälje, Sweden
| | - Camilla I Svensson
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
7
|
Neurotrophic and antioxidant effects of silymarin comparable to 4-methylcatechol in protection against gentamicin-induced ototoxicity in guinea pigs. Pharmacol Rep 2014; 67:317-25. [PMID: 25712657 DOI: 10.1016/j.pharep.2014.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 09/14/2014] [Accepted: 10/06/2014] [Indexed: 11/21/2022]
Abstract
BACKGROUND Despite that gentamicin is a very effective aminoglycoside, its potential ototoxicity which is of irreversible nature makes a challenge and limitation for its use. This study was designed to investigate possible neurotrophic and antioxidant effects of silymarin comparable to 4-methylcatechol in protection against gentamicin-induced ototoxicity. METHODS AND RESULTS Twenty pigmented guinea pigs were divided into four equal groups, where group I served as normal control group. The other groups received gentamicin (120 mg/kg/day, ip) for 19 days where group II given vehicle of 1% CMC, group III and group IV were pre-treated 2h before gentamicin by 4-methylcatechol (10 μg/kg, ip) and silymarin (100mg/kg, oral gavage), respectively. The main findings indicated that silymarin exhibited restoration of nerve growth factor (NGF) levels and increased tropomyosin-related kinase receptors-A (Trk-A) m-RNA expression in cochlear tissue and preservation of hair cells of organ of Corti by scanning electron microscopy (SEM) with significant decrease in auditory brainstem response (ABR) threshold compared to 4-methylcatechol. Only silymarin caused significant amelioration in oxidative stress state by reducing malondialdehyde (MDA) levels and increasing catalase activity. CONCLUSIONS Silymarin exerts superiority over 4-methylcatechol when recommended as protective agent against gentamicin ototoxicity based on its efficient neurotrophic and antioxidant activities.
Collapse
|
8
|
Siniscalco D, Giordano C, Rossi F, Maione S, de Novellis V. Role of neurotrophins in neuropathic pain. Curr Neuropharmacol 2012; 9:523-9. [PMID: 22654713 PMCID: PMC3263449 DOI: 10.2174/157015911798376208] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 07/06/2010] [Accepted: 07/06/2010] [Indexed: 11/27/2022] Open
Abstract
Neurotrophins (NTs) belong to a family of structurally and functionally related proteins, they are the subsets of neurotrophic factors. Neurotrophins are responsible for diverse actions in the developing peripheral and central nervous systems. They are important regulators of neuronal function, affecting neuronal survival and growth. They are able to regulate cell death and survival in development as well as in pathophysiologic states. NTs and their receptors are expressed in areas of the brain that undergo plasticity, indicating that they are able to modulate synaptic plasticity. Recently, neurotrophins have been shown to play significant roles in the development and transmission of neuropathic pain. Neuropathic pain is initiated by a primary lesion or dysfunction in the nervous system. It has a huge impact on the quality of life. It is debilitating and often has an associated degree of depression that contributes to decreasing human well being. Neuropathic pain ranks at the first place for sanitary costs. Neuropathic pain treatment is extremely difficult. Several molecular pathways are involved, making it a very complex disease. Excitatory or inhibitory pathways controlling neuropathic pain development show altered gene expression, caused by peripheral nerve injury. At present there are no valid treatments over time and neuropathic pain can be classified as an incurable disease. Nowadays, pain research is directing towards new molecular methods. By targeting neurotrophin molecules it may be possible to provide better pain control than currently available.
Collapse
Affiliation(s)
- Dario Siniscalco
- Department of Experimental Medicine, Division of Pharmacology "L. Donatelli", Second University of Naples, Via S. Maria di Costantinopoli, 16 - 80138 Naples, Italy
| | | | | | | | | |
Collapse
|
9
|
Mills CD, Nguyen T, Tanga FY, Zhong C, Gauvin DM, Mikusa J, Gomez EJ, Salyers AK, Bannon AW. Characterization of nerve growth factor-induced mechanical and thermal hypersensitivity in rats. Eur J Pain 2012; 17:469-79. [PMID: 22915527 DOI: 10.1002/j.1532-2149.2012.00202.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2012] [Indexed: 11/08/2022]
Abstract
BACKGROUND Injection of nerve growth factor (NGF) produces mechanical and thermal hypersensitivity in rodents and humans. Treatment with sequestering antibodies demonstrates the importance of NGF in various pain states, with efficacy seen in a number of animal pain models and in painful human conditions. However, these phenomena have not been evaluated in the context of using NGF-induced hypersensitivities as a model of pain. METHODS NGF-induced behaviours were characterized using von Frey filament, pinprick and thermal endpoints and then pharmacologically evaluated with known reference agents. RESULTS Intraplantar NGF injection produced a dose-dependent increase in thermal sensitivity that lasted through 24 h post-injection and an immediate long-lasting (2 week) increase in mechanical sensitivity at the injection site, with no effects detected at secondary sites. NGF-induced mechanical sensitivity was pharmacologically characterized at 4 h and 1 week post-NGF injection. The nonsteroidal anti-inflammatory drugs (NSAIDs), celecoxib and diclofenac, were minimally effective against both thermal and mechanical endpoints. Gabapenitn and duloxetine were only moderately effective against thermal and mechanical hypersensitivity. Morphine was effective against thermal and mechanical endpoints at every time point examined. Treatment with the transient receptor potential vanilloid 1 (TRPV1) antagonist A-784168 partially attenuated NGF-induced thermal and mechanical sensitivity at all time points examined. CONCLUSIONS The results reported here suggest that effects of NGF on thermal and mechanical sensitivity in rats are similar to those reported in human and are partially driven by TRPV1. The rat NGF model may serve as a potential translational model for exploring the effects of novel analgesic agents.
Collapse
Affiliation(s)
- C D Mills
- Department of Neuroscience, Abbott Laboratories, Abbott Park, IL, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Nerve Growth Factor (NGF) was initially studied for its role as a key player in the regulation of peripheral innervations. However, the successive finding of its release in the bloodstream of male mice following aggressive encounters and its presence in the central nervous system led to the hypothesis that variations in brain NGF levels, caused by psychosocial stressor, and the related alterations in emotionality, could be functional to the development of proper strategies to cope with the stressor itself and thus to survive. Years later this vision is still relevant, and the body of evidence on the role of NGF has been strengthened and expanded from trophic factor playing a role in brain growth and differentiation to a much more complex messenger, involved in psychoneuroendocrine plasticity.
Collapse
|
11
|
Te AE. A study to investigate tanezumab in patients with interstitial cystitis/painful bladder syndrome. Curr Urol Rep 2011; 12:245-6. [PMID: 21559849 DOI: 10.1007/s11934-011-0194-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
12
|
Mantyh PW, Koltzenburg M, Mendell LM, Tive L, Shelton DL. Antagonism of nerve growth factor-TrkA signaling and the relief of pain. Anesthesiology 2011; 115:189-204. [PMID: 21602663 PMCID: PMC3121917 DOI: 10.1097/aln.0b013e31821b1ac5] [Citation(s) in RCA: 244] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Nerve growth factor (NGF) was originally discovered as a neurotrophic factor essential for the survival of sensory and sympathetic neurons during development. However, in the adult NGF has been found to play an important role in nociceptor sensitization after tissue injury. The authors outline mechanisms by which NGF activation of its cognate receptor, tropomyosin-related kinase A receptor, regulates a host of ion channels, receptors, and signaling molecules to enhance acute and chronic pain. The authors also document that peripherally restricted antagonism of NGF-tropomyosin-related kinase A receptor signaling is effective for controlling human pain while appearing to maintain normal nociceptor function. Understanding whether there are any unexpected adverse events and how humans may change their behavior and use of the injured/degenerating tissue after significant pain relief without sedation will be required to fully appreciate the patient populations that may benefit from these therapies targeting NGF.
Collapse
Affiliation(s)
- Patrick W Mantyh
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA.
| | | | | | | | | |
Collapse
|
13
|
Sex differences in neuropeptide content and release from rat dental pulp. J Endod 2011; 37:1098-101. [PMID: 21763901 DOI: 10.1016/j.joen.2011.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 03/15/2011] [Accepted: 03/17/2011] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Studies to examine sex differences in response to pain have suggested that females exhibit lower threshold responses to painful stimuli and that threshold response varies greatly at different stages of the menstrual cycle. Additional studies suggest that sex differences may be caused by societal sex roles or differences in anxiety responses by men and women. OBJECTIVE The purpose of this study was to evaluate biologically evident sex differences in male and female rats chronically treated with a systemic algogen, the nerve growth factor (NGF), by measuring neuropeptides (calcitonin gene-related peptide) content and release from isolated dental pulp. METHODS Rats were injected subcutaneously every other day with either murine NGF (1 mg/kg) or vehicle for 7 or 13 days. Isolated incisor pulp tissue was evaluated from these male and female rats (n = 96). Capsaicin-evoked neurosecretion of CGRP and tissue content were measured using a previously validated radioimmunoassay. RESULTS Dental pulp from female rats at 7 days showed significantly increased capsaicin-evoked immunoreactive CGRP release (>50% increase) compared with tissue from male rats. After 13 days, this release was significantly increased only in NGF-treated female rats (3-fold increase) when compared with control females or both male groups. The CGRP content in tissue from both female groups was also significantly increased after 7 days of treatment (>3 fold), but after 13 days this content was only significantly increased in tissue from NGF-treated female rats (P = .0001). CONCLUSIONS These data suggest that sex differences affect the role of NGF in the modulation of inflammation through the regulation of peripheral neuropeptide release and content.
Collapse
|
14
|
|
15
|
Cirulli F, Alleva E. The NGF saga: from animal models of psychosocial stress to stress-related psychopathology. Front Neuroendocrinol 2009; 30:379-95. [PMID: 19442684 DOI: 10.1016/j.yfrne.2009.05.002] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Revised: 04/17/2009] [Accepted: 05/04/2009] [Indexed: 01/15/2023]
Abstract
The role of the neurotrophins Nerve Growth Factor (NGF) and Brain-Derived Neurotrophic Factor (BDNF) has been expanding over the last years from trophic factors involved in brain growth and differentiation, to much more complex messengers, involved in psycho-neuro-endocrine adaptations. Much of this research stems from a series of studies inspired by the life-long work of the Nobel laureate Rita Levi-Montalcini. A new field of research started when NGF was found to be released in the bloodstream as a result of psychosocial stressors in male mice. Subsequent studies have shown that, in humans, highly arousing situations also result in increased blood levels of NGF, underlying the unique role of this neurotrophin, compared to other neuroendocrine effectors, and its sensitivity to environmental variables endowed by a social nature. Data are reviewed to support the hypothesis that this neurotrophic factor, together with BDNF, could be involved in the neurobiological changes underlying physiological and pathological reactions to stress that can result in increased vulnerability to disease in humans, including risk for anxiety disorders, or in the complex pathophysiology associated with mood disorders. Indeed, numerous data indicate that neurotrophins are present in brain hypothalamic areas involved in the regulation of hypothalamic-pituitary-adrenal axis, circadian rhythms and metabolism. In addition, there is now evidence that, in addition to the nervous system, neurotrophins exert their effects in various tissue compartments as they are produced by a variety of non-neuronal cell types such as endocrine and immune cells, adipocytes, endothelial cells, keratinocytes, thus being in a position to coordinate brain and body reactions to external challenges. Aim of this review is to discuss the evidence suggesting a role for neurotrophins as multifunctional signaling molecules activated during allostatic responses to stressful events and their involvement in the complex pathophysiology underlying stress-related psychopathology.
Collapse
Affiliation(s)
- Francesca Cirulli
- Section of Behavioural Neurosciences, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità Rome, Viale Regina Elena 299, I-00161 Roma, Italy.
| | | |
Collapse
|
16
|
Stein C, Clark JD, Oh U, Vasko MR, Wilcox GL, Overland AC, Vanderah TW, Spencer RH. Peripheral mechanisms of pain and analgesia. ACTA ACUST UNITED AC 2008; 60:90-113. [PMID: 19150465 DOI: 10.1016/j.brainresrev.2008.12.017] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2008] [Indexed: 12/23/2022]
Abstract
This review summarizes recent findings on peripheral mechanisms underlying the generation and inhibition of pain. The focus is on events occurring in peripheral injured tissues that lead to the sensitization and excitation of primary afferent neurons, and on the modulation of such mechanisms. Primary afferent neurons are of particular interest from a therapeutic perspective because they are the initial generator of noxious impulses traveling towards relay stations in the spinal cord and the brain. Thus, if one finds ways to inhibit the sensitization and/or excitation of peripheral sensory neurons, subsequent central events such as wind-up, sensitization and plasticity may be prevented. Most importantly, if agents are found that selectively modulate primary afferent function and do not cross the blood-brain-barrier, centrally mediated untoward side effects of conventional analgesics (e.g. opioids, anticonvulsants) may be avoided. This article begins with the peripheral actions of opioids, turns to a discussion of the effects of adrenergic co-adjuvants, and then moves on to a discussion of pro-inflammatory mechanisms focusing on TRP channels and nerve growth factor, their signaling pathways and arising therapeutic perspectives.
Collapse
Affiliation(s)
- Christoph Stein
- Department of Anesthesiology and Critical Care Medicine, Charité Campus Benjamin Franklin, Freie Universität Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Xu P, Hall AK. The role of activin in neuropeptide induction and pain sensation. Dev Biol 2006; 299:303-9. [PMID: 16973148 DOI: 10.1016/j.ydbio.2006.08.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 08/05/2006] [Accepted: 08/12/2006] [Indexed: 11/16/2022]
Abstract
Signals from target tissues play critical roles in the functional differentiation of neuronal cells, and in their subsequent adaptations to peripheral changes in the adult. Sensory neurons in the dorsal root ganglia (DRG) provide an excellent model system for the study of signals that regulate the development of neuronal diversity. DRG have been well characterized and contain both neurons that convey information from muscles about limb position, as well as other neurons that provide sensations from skin about pain information. Sensory neurons involved in pain sensation can be distinguished physiologically and antigenically, and one hallmark characteristic is that these neurons contain neuropeptides important for their functions. The transforming growth factor (TGF) beta family member activin A has recently been implicated in neural development and response to injury. During sensory neuron development, peripheral target tissues containing activin or activin itself can regulate pain neuropeptide expression. Long after development has ceased, skin target tissues retain the capacity to signal neurons about changes or injury, to functionally refine synapses. This review focuses on the role of activin as a target-derived differentiative factor in neural development that has additional roles in response to cutaneous injuries in the adult.
Collapse
Affiliation(s)
- Pin Xu
- Department of Neuroscience, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | | |
Collapse
|
18
|
Moalem G, Tracey DJ. Immune and inflammatory mechanisms in neuropathic pain. ACTA ACUST UNITED AC 2006; 51:240-64. [PMID: 16388853 DOI: 10.1016/j.brainresrev.2005.11.004] [Citation(s) in RCA: 577] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Revised: 11/16/2005] [Accepted: 11/17/2005] [Indexed: 12/22/2022]
Abstract
Tissue damage, inflammation or injury of the nervous system may result in chronic neuropathic pain characterised by increased sensitivity to painful stimuli (hyperalgesia), the perception of innocuous stimuli as painful (allodynia) and spontaneous pain. Neuropathic pain has been described in about 1% of the US population, is often severely debilitating and largely resistant to treatment. Animal models of peripheral neuropathic pain are now available in which the mechanisms underlying hyperalgesia and allodynia due to nerve injury or nerve inflammation can be analysed. Recently, it has become clear that inflammatory and immune mechanisms both in the periphery and the central nervous system play an important role in neuropathic pain. Infiltration of inflammatory cells, as well as activation of resident immune cells in response to nervous system damage, leads to subsequent production and secretion of various inflammatory mediators. These mediators promote neuroimmune activation and can sensitise primary afferent neurones and contribute to pain hypersensitivity. Inflammatory cells such as mast cells, neutrophils, macrophages and T lymphocytes have all been implicated, as have immune-like glial cells such as microglia and astrocytes. In addition, the immune response plays an important role in demyelinating neuropathies such as multiple sclerosis (MS), in which pain is a common symptom, and an animal model of MS-related pain has recently been demonstrated. Here, we will briefly review some of the milestones in research that have led to an increased awareness of the contribution of immune and inflammatory systems to neuropathic pain and then review in more detail the role of immune cells and inflammatory mediators.
Collapse
Affiliation(s)
- Gila Moalem
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | | |
Collapse
|
19
|
Xu P, Van Slambrouck C, Berti-Mattera L, Hall AK. Activin induces tactile allodynia and increases calcitonin gene-related peptide after peripheral inflammation. J Neurosci 2005; 25:9227-35. [PMID: 16207882 PMCID: PMC6725762 DOI: 10.1523/jneurosci.3051-05.2005] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP) is a sensory neuropeptide important in inflammatory pain that conveys pain information centrally and dilates blood vessels peripherally. Previous studies indicate that activin A increases CGRP-immunoreactive (IR) sensory neurons in vitro, and following wound, activin A protein increases in the skin and more neurons have detectable CGRP expression in the innervating dorsal root ganglion (DRG). These data suggest some adult sensory neurons respond to activin A or other target-derived factors with increased neuropeptide expression. This study was undertaken to test whether activin contributes to inflammatory pain and increased CGRP and to learn which neurons retained plasticity. After adjuvant-induced inflammation, activin mRNA, but not NGF or glial cell line-derived neurotrophic factor, increased in the skin. To examine which DRG neurons increased CGRP immunoreactivity, retrograde tracer-labeled cutaneous neurons were characterized after inflammation. The proportion and size of tracer-labeled DRG neurons with detectable CGRP increased after inflammation. One-third of CGRP-IR neurons that appear after inflammation also had isolectin B4 binding, suggesting that some mechanoreceptors became CGRP-IR. In contrast, the increased proportion of CGRP-IR neurons did not appear to come from RT97-IR neurons. To learn whether central projections were altered after inflammation, CGRP immunoreactivity in the protein kinase Cgamma-IR lamina IIi was quantified and found to increase. Injection of activin A protein alone caused robust tactile allodynia and increased CGRP in the DRG. Together, these data support the hypothesis that inflammation and skin changes involving activin A cause some sensory neurons to increase CGRP expression and pain responses.
Collapse
Affiliation(s)
- Pin Xu
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
20
|
Yeomans DC, Levinson SR, Peters MC, Koszowski AG, Tzabazis AZ, Gilly WF, Wilson SP. Decrease in inflammatory hyperalgesia by herpes vector-mediated knockdown of Nav1.7 sodium channels in primary afferents. Hum Gene Ther 2005; 16:271-7. [PMID: 15761266 DOI: 10.1089/hum.2005.16.271] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Induction of peripheral inflammation increases the expression of the Nav1.7 sodium channel in sensory neurons, potentially increasing their excitability. Peripheral inflammation also produces hyperalgesia in humans and an increase in nociceptive responsiveness in animals. To test the relationship between these two phenomena we applied a recombinant herpes simplex-based vector to the hindpaw skin of mice, which encoded both green fluorescent protein (GFP) as well as an antisense sequence to the Nav1.7 gene. The hindpaw was subsequently injected with complete Freund's adjuvant to induce robust inflammation. Application of the vector, but not a control vector encoding only GFP, prevented an increase in Nav1.7 expression in GFP-positive neurons and prevented development of hyperalgesia in both C and Adelta thermonociceptive tests. These results provide clear evidence of the involvement of an increased expression of the Nav1.7 channel in nociceptive neurons in the development of inflammatory hyperalgesia.
Collapse
Affiliation(s)
- D C Yeomans
- Department of Anesthesia and Stanford Pain and Analgesia Research Center, Stanford University, Stanford, CA 94305-5117, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Zuch CL, David D, Ujhelyi L, Hudson JL, Gerhardt GA, Kaplan PL, Bickford PC. Beneficial effects of intraventricularly administered BMP-7 following a striatal 6-hydroxydopamine lesion. Brain Res 2004; 1010:10-6. [PMID: 15126112 DOI: 10.1016/j.brainres.2003.12.058] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2003] [Indexed: 11/25/2022]
Abstract
The present study was undertaken to investigate the effects of bone morphogenetic protein-7 (BMP-7), also named osteogenic protein-1 (OP-1), on the progression of a striatal 6-hydroxydopamine (6-OHDA) lesion. BMP-7, a member of the transforming growth factor-beta (TGF-beta) superfamily of proteins, has been shown to have protective effects in other animal models of neuronal damage. In this study, male Fischer 344 rats received striatal 6-OHDA lesions followed 1 week later by an intraventricular dose of BMP-7. No significant effect of BMP-7 treatment on spontaneous locomotor activity was observed, however BMP-7 significantly increased the density of tyrosine hydroxylase (TH) immunoreactivity (TH-ir) in the substantia nigra (SN) pars compacta, in the lesioned hemisphere [31.7+/-5.2 (optical density (O.D.) arbitrary units) control vs. 50.2+/-4.3 O.D. BMP-7-treated; p<0.05]. Interestingly, BMP-7 significantly increased TH-ir in the SN of the non-lesioned hemisphere (pars reticulata: 14.8+/-1.19 O.D. control vs. 36+/-2.6 O.D. BMP-7-treated, p<0.05; pars compacta: 29.0+/-4.9 O.D. control vs. 64.4+/-6.9 O.D. BMP-7-treated, p<0.001). A significant increase in DA concentration in the contralateral, non-lesioned hemisphere was also noted (113.2 ng/g control vs. 198.2 ng/g BMP-7-treated, p<0.01). In contrast to other intraventricularly administered neurotrophic factors, BMP-7 was not associated with an increase in the sensitivity to pain. These results suggest that BMP-7 is able to act as a dopaminotrophic agent without unwanted side effects and as such may be a useful pharmacological tool in the treatment of Parkinson's disease in humans.
Collapse
Affiliation(s)
- Christina L Zuch
- Department of Pharmacology, University of Colorado Health Sciences Center, Denver, CO, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Nerve growth factor (NGF) was discovered 50 years ago as a molecule that promoted the survival and differentiation of sensory and sympathetic neurons. Its roles in neural development have been characterized extensively, but recent findings point to an unexpected diversity of NGF actions and indicate that developmental effects are only one aspect of the biology of NGF. This article considers expanded roles for NGF that are associated with the dynamically regulated production of NGF and its receptors that begins in development, extends throughout adult life and aging, and involves a surprising variety of neurons, glia, and nonneural cells. Particular attention is given to a growing body of evidence that suggests that among other roles, endogenous NGF signaling subserves neuroprotective and repair functions. The analysis points to many interesting unanswered questions and to the potential for continuing research on NGF to substantially enhance our understanding of the mechanisms and treatment of neurological disorders.
Collapse
Affiliation(s)
- M V Sofroniew
- Department of Neurobiology and Brain Research Institute, University of California Los Angeles, Los Angeles, California 90095-1763, USA.
| | | | | |
Collapse
|
23
|
Abstract
Adult male mouse submaxillary glands served as the preferred starting material for the isolation of the nerve growth factor (NGF) proteins in most of the isolation studies done. Two types of NGF proteins were isolated from extracts of the gland, a high-molecular-weight 7S NGF complex and a low-molecular-weight protein variously called NGF, betaNGF, or 2.5S NGF. The latter, which mediated all known biological functions of NGF, were closely related forms of a basic NGF dimer in which the N and C termini of two monomers (chains) were modified by proteolytic enzymes to different extents with no effect on biological activity. The betaNGF dimer showed a novel protein structure in which the two chains interacted non-covalently over a wide surface. Correspondingly, the betaNGF dimer was found to be unusually stable and the form through which NGFs actions were mediated at physiological concentrations. The betaNGF dimer was one of three subunits in 7S NGF; the other two were the gamma subunit, an arginine esteropeptidase or kallikrein, and the alpha subunit, an inactive kallikrein. Two zinc ions were also present in the complex and contributed greatly to its stability. There was much debate about whether 7S NGF was a specific protein complex of interacting subunits and, if so, what functions it might play in the biology of NGF. Observations of the inhibition of the enzyme activity of the gamma subunit and of the biological activity of betaNGF in 7S NGF were important in determining that 7S NGF was a naturally occurring complex and the sole source of NGF in the gland extract or in saliva. Specific interactions between the active site of the gamma subunit and the C-terminal arginine residues of the NGF chains, confirmed in the three-dimensional structure of 7S NGF, suggested a role for the gamma subunit in pro-NGF processing during the assembly of 7S NGF. In spite of the detailed knowledge of 7S NGF structure, no information on the role of this complex in the neurobiology of NGF has emerged. With the exception of the submaxillary gland of an African rodent, no other source of NGF has been convincingly shown to synthesize the alpha and gamma subunits, and they may well be irrelevant to NGFs actions.
Collapse
Affiliation(s)
- E M Shooter
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94305-5125, USA.
| |
Collapse
|
24
|
Goettl VM, Lindsey AE, Neff NH, Hadjiconstantinou M. GM1 ganglioside restores abnormal responses to acute thermal and mechanical stimuli in aged rats. Brain Res 2000; 858:380-5. [PMID: 10708690 DOI: 10.1016/s0006-8993(00)01960-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We investigated the effect of aging on the responses to thermal and mechanical stimuli in rats. Young (3-5 months old) and aged (22-24 months old) male Sprague-Dawley rats were tested in the hot plate, high- and low-intensity radiant heat tail flick, and von Frey hair assays. Compared to young rats, aged rats displayed longer latencies in the hot plate and the high-intensity tail flick assays (hypoalgesia), but there was no difference in the low-intensity tail flick assay. In addition, aged rats had decreased thresholds to mechanical stimuli produced by von Frey hairs compared with young rats (mechanical allodynia). Administration of GM1 ganglioside, 30 mg/kg, i.p., once daily for 30 days, to aged rats partially restored the responses in the hot plate and von Frey hair assays. GM1 had no effect on the altered responses in the tail flick test in aged rats, and in general, had no effect on any sensory modality tested in young rats.
Collapse
Affiliation(s)
- V M Goettl
- Department of Pharmacology, The Ohio State University College of Medicine and Public Health, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
25
|
Laudanna A, Nogueira MI, Mariano M. Expression of Fos protein in the rat central nervous system in response to noxious stimulation: effects of chronic inflammation of the superior cervical ganglion. Braz J Med Biol Res 1998; 31:847-50. [PMID: 9698833 DOI: 10.1590/s0100-879x1998000600019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The aim of this study was to investigate the possible interactions between the nociceptive system, the sympathetic system and the inflammatory process. Thus, the superior cervical ganglion of rats was submitted to chronic inflammation and Fos expression was used as a marker for neuronal activity throughout central neurons following painful peripheral stimulation. The painful stimulus consisted of subcutaneously injected formalin applied to the supra-ocular region. Fos-positive neurons were identified by conventional immunohistochemical techniques, and analyzed from the obex through the cervical levels of the spinal cord. In the caudal sub-nucleus of the spinal trigeminal nuclear complex, the number of Fos-positive neurons was much higher in rats with inflammation of the superior cervical ganglion than in control rats, either sham-operated or with saline applied to the ganglion. There was a highly significant difference in the density of Fos-positive neurons between the inflamed and control groups. No significant difference was found between control groups. These results suggest that the inflammation of the superior cervical ganglion generated an increased responsiveness to painful stimuli, which may have been due to a diminished sympathetic influence upon the sensory peripheral innervation.
Collapse
Affiliation(s)
- A Laudanna
- Departamento de Fisiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brasil.
| | | | | |
Collapse
|
26
|
Levi-Montalcini R, Skaper SD, Dal Toso R, Petrelli L, Leon A. Nerve growth factor: from neurotrophin to neurokine. Trends Neurosci 1996; 19:514-20. [PMID: 8931279 DOI: 10.1016/s0166-2236(96)10058-8] [Citation(s) in RCA: 449] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Nerve growth factor (NGF) is largely known as a target-derived factor responsible for the survival and maintenance of the phenotype of specific subsets of peripheral neurones and basal forebrain cholinergic nuclei during development and maturation. However, NGF also exerts a modulatory role on sensory, nociceptive nerve physiology during adulthood that appears to correlate with hyperalgesic phenomena occurring in tissue inflammation. Other NGF-responsive cells are now recognized as belonging to the haemopoietic-immune system and to populations in the brain involved in neuroendocrine functions. The concentration of NGF is elevated in a number of inflammatory and autoimmune states in conjunction with an increased accumulation of mast cells. Mast cells and NGF appear to be involved in neuroimmune interactions and tissue inflammation, with NGF acting as a general 'alert' molecule capable of recruiting and priming tissue defence processes following insult as well as systemic defensive mechanisms. Moreover, mast cells themselves produce NGF, suggesting that alterations in normal mast cell behaviours can provoke maladaptive neuroimmune tissue responses whose consequences could have profound implications in inflammatory disease states. This review discusses recent discoveries involving novel and diverse biological activities of this fascinating molecule.
Collapse
Affiliation(s)
- R Levi-Montalcini
- Institute of Neurobiology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | | | | | | | | |
Collapse
|
27
|
Falcini F, Matucci Cerinic M, Lombardi A, Generini S, Pignone A, Tirassa P, Ermini M, Lepore L, Partsch G, Aloe L. Increased circulating nerve growth factor is directly correlated with disease activity in juvenile chronic arthritis. Ann Rheum Dis 1996; 55:745-8. [PMID: 8984940 PMCID: PMC1010293 DOI: 10.1136/ard.55.10.745] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To determine the circulating serum concentrations of nerve growth factor (NGF) and compare them with indices of disease activity in juvenile chronic arthritis. METHODS NGF concentrations were evaluated with a two site immunoenzymatic assay (ELISA), in 17 children with systemic, 39 with polyarticular, and 24 with pauciarticular onset juvenile chronic arthritis. Each subset was divided according to different variables, appropriate to each subset, reflecting active and inactive disease. RESULTS NGF concentrations were significantly higher in children with systemic [254 (SD 256.1) pg ml-1; P < 0.001], polyarticular [165.2 (300.8) pg ml-1; P < 0.05], and pauciarticular [106.8 (111.8) pg ml-1; P < 0.005] onset juvenile chronic arthritis than in controls. In all subsets, NGF concentrations were higher in the active than in the inactive phase of the disease. A significant direct correlation between NGF concentrations and erythrocyte sedimentation rate was found both in the systemic and in the polyarticular onset juvenile chronic arthritis. CONCLUSIONS The increase in NGF concentrations in all juvenile chronic arthritis subsets and the correlation with disease activity suggest that NGF may take an active part in joint inflammation.
Collapse
Affiliation(s)
- F Falcini
- Department of Paediatrics, University of Florence, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Damage to peripheral nerves often results in pain and hyperalgesia. We suggest that nerve damage causes an inflammatory response in which cells associated with the nerve release inflammatory mediators such as eicosanoids; these mediators may contribute to the hyperalgesia which results from nerve injury. The cell types most likely to be responsible include macrophages and postganglionic sympathetic neurones. A better understanding of the mechanisms involved should lead to improved therapies for neuropathic pain.
Collapse
Affiliation(s)
- D J Tracey
- School of Anatomy, University of New South Wales, Sydney, Australia
| | | |
Collapse
|
29
|
Safieh-Garabedian B, Poole S, Allchorne A, Winter J, Woolf CJ. Contribution of interleukin-1 beta to the inflammation-induced increase in nerve growth factor levels and inflammatory hyperalgesia. Br J Pharmacol 1995; 115:1265-75. [PMID: 7582555 PMCID: PMC1908795 DOI: 10.1111/j.1476-5381.1995.tb15035.x] [Citation(s) in RCA: 474] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
1. Peripheral inflammation is associated with the local production of neuroactive inflammatory cytokines and growth factors. These may contribute to inflammatory pain and hyperalgesia by directly or indirectly altering the function or chemical phenotype of responsive primary sensory neurones. 2. To investigate this, inflammation was produced by the intraplantar injection of complete Freund's adjuvant (CFA) in adult rats. This resulted in a significant elevation in interleukin-1 beta (IL-1 beta) and nerve growth factor (NGF) levels in the inflamed tissue and of the peptides, substance P and calcitonin gene-related peptide (CGRP) in the L4 dorsal root ganglion 48 h post CFA injection. 3. The effects of a steroidal (dexamethasone) and a non-steroidal (indomethacin) anti-inflammatory drug on the levels of NGF and IL-1 beta in inflamed tissue were investigated and compared with alterations in behavioural hyperalgesia and neuropeptide expression in sensory neurones. 4. Systemic dexamethasone (120 micrograms kg-1 per day starting the day before the CFA injection) had no effect on the inflammatory hyperalgesia. When the dose was administered 3 times daily, a reduction in mechanical and to a lesser extent thermal sensitivity occurred. Indomethacin at 2 mg kg-1 daily (i.p.) had no effect on the hyperalgesia and a dose of 4 mg kg-1 daily was required to reduce significantly mechanical and thermal hypersensitivity. 5. The increase in NGF produced by the CFA inflammation was prevented by both dexamethasone and indomethacin, but only at the higher dose levels. Dexamethasone at the lower and higher dose regimes diminished the upregulation of IL-1 beta whereas indomethacin had an effect only at the higher dose. 6. The increase in SP and CGRP levels produced by the CFA inflammation was prevented by dexamethasone and indomethacin at the lower and higher dose regimes. 7. Intraplantar injections of IL-1 beta (0.01, 0.1 and 1 ng) produced a brief (6 h) thermal hyperalgesia and an elevation in cutaneous NGF levels which was prevented by pretreatment with human recombinant IL-1 receptor antagonist (IL-1 ra) (0.625 microgram, i.v.). The thermal hyperalgesia but not the NGF elevation produced by intraplantar IL-1 beta (1 ng) was prevented by administration of a polyclonal neutralizing anti-NGF serum. 8. IL-1 ra significantly reduced the mechanical hyperalgesia produced by CFA for 6 h after administration as well as the CFA-induced elevation in NGF levels. Anti-NGF pretreatment substantially reduced CFA-induced mechanical and thermal hyperalgesia without reducing the elevation in IL-1 beta. 9. Intraplantar NGF (0.02, 0.2 and 2 microg) injections produced a short lasting thermal and mechanical hyperalgesia but did not change IL-1beta levels in the hindpaw skin.10. Our results demonstrate that IL-1beta contributes to the upregulation of NGF during inflammation and that NGF has a major role in the production of inflammatory pain hypersensitivity.
Collapse
|
30
|
Abstract
The sympathetic nervous system serves not only to regulate involuntary functions, but also appears to play an important part in modulating sensory processing. While studies in animal models of neuropathic pain and clinical observations point to a role of the sympathetic nervous system in certain chronic pain states, the function of the sympathetics in postoperative pain and inflammation is debatable. Behavioural studies in rats point to a contribution of the sympathetic postganglionic terminal in the hyperalgesia of cutaneous inflammation and the severity of arthritis. An indirect effect of noradrenaline and inflammatory mediators via the release of prostaglandins has been postulated. Neurophysiological studies of nociceptors in rats and psychophysical studies in humans have failed to provide confirmatory evidence for the role of the sympathetic efferents in inflammatory pain and hyperalgesia. The clinical significance of the potential interaction of the sympathetic nervous system and the somatic afferent system needs further investigation.
Collapse
Affiliation(s)
- S N Raja
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21281-5354, USA
| |
Collapse
|
31
|
Leon A, Buriani A, Dal Toso R, Fabris M, Romanello S, Aloe L, Levi-Montalcini R. Mast cells synthesize, store, and release nerve growth factor. Proc Natl Acad Sci U S A 1994; 91:3739-43. [PMID: 8170980 PMCID: PMC43657 DOI: 10.1073/pnas.91.9.3739] [Citation(s) in RCA: 451] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Mast cells and nerve growth factor (NGF) have both been reported to be involved in neuroimmune interactions and tissue inflammation. In many peripheral tissues, mast cells interact with the innervating fibers. Changes in the behaviors of both of these elements occur after tissue injury/inflammation. As such conditions are typically associated with rapid mast cell activation and NGF accumulation in inflammatory exudates, we hypothesized that mast cells may be capable of producing NGF. Here we report that (i) NGF mRNA is expressed in adult rat peritoneal mast cells; (ii) anti-NGF antibodies clearly stain vesicular compartments of purified mast cells and mast cells in histological sections of adult rodent mesenchymal tissues; and (iii) medium conditioned by peritoneal mast cells contains biologically active NGF. Mast cells thus represent a newly recognized source of NGF. The known actions of NGF on peripheral nerve fibers and immune cells suggest that mast cell-derived NGF may control adaptive/reactive responses of the nervous and immune systems toward noxious tissue perturbations. Conversely, alterations in normal mast cell behaviors may provoke maladaptive neuroimmune tissue responses whose consequences could have profound implications in inflammatory disease states, including those of an autoimmune nature.
Collapse
Affiliation(s)
- A Leon
- Researchlife, Ospedale Civile, Castelfranco Veneto, Italy
| | | | | | | | | | | | | |
Collapse
|
32
|
Alleva E, Aloe L, Bigi S. An updated role for nerve growth factor in neurobehavioural regulation of adult vertebrates. Rev Neurosci 1993; 4:41-62. [PMID: 7952382 DOI: 10.1515/revneuro.1993.4.1.41] [Citation(s) in RCA: 57] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Increasing attention has been focused on the role(s) of nerve growth factor (NGF) in neurobehavioural regulations of adult vertebrates. This interest springs from the emerging evidence that NGF is a "regulator" of physiological processes belonging to the three main homeostatic systems: the nervous, immune and endocrine systems. In fact, the spectrum of action of the NGF molecule is not restricted to neuronal cell types (central basal forebrain; peripheral sensory and sympathetic neurons) but extends also to nonneuronal cells. In mice intermale aggressive behaviour enhances serum NGF levels and promotes its synthesis in some hypothalamic areas. Other types of social events are able to cause NGF release, particularly under stress conditions. The achievement of a social role (dominant vs subordinate) is due to a functional loop involving salivary NGF release-->enhanced production of adrenal hormones-->submissive behaviour-->NGF release. In humans, plasma platelet-derived growth factor (PDGF) increases following mental stress. The aim of this review is to give an updated survey on NGF roles in neurobehavioural regulations of adult animals.
Collapse
Affiliation(s)
- E Alleva
- Section of Behavioural Pathophysiology, Istituto Superiore di Sanità, Rome, Italy
| | | | | |
Collapse
|
33
|
Affiliation(s)
- E Weihe
- Department of Anatomy, Johannes-Gutenberg-Universität Mainz, Germany
| |
Collapse
|