1
|
Farham F, Onan D, Martelletti P. Non-Migraine Head Pain and Botulinum Toxin. Toxins (Basel) 2024; 16:431. [PMID: 39453207 PMCID: PMC11511419 DOI: 10.3390/toxins16100431] [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: 08/24/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024] Open
Abstract
Botulinum toxin A (BT-A), a potential neurotoxin produced by the bacterium Clostridium botulinum, is known for its ability to prevent the release of acetylcholine at the neuromuscular synapse, leading to temporary muscle paralysis. BT-A is used for a wide range of therapeutic applications. Several studies have shown mechanisms beyond the inhibition of acetylcholine release for pain control. BT-A inhibits the release of neurotransmitters associated with pain and inflammation, such as glutamate, CGRP, and substance P. Additionally, it would be effective in nerve entrapment leading to neuronal hypersensitivity, which is known as a new pathogenesis of painful conditions. BT-A has been applied to the treatment of a wide variety of neurological disorders. Since 2010, BT-A application has been approved and widely used as a chronic migraine prophylaxis. Moreover, due to its effects on pain through sensory modulation, it may also be effective for other headaches. Several studies using BT-A, at different doses and administration sites for headaches, have shown beneficial effects on frequency and severity. In this review, we provide an overview of using BT-A to treat primary and secondary headache disorders.
Collapse
Affiliation(s)
- Fatemeh Farham
- Department of Headache, Iranian Centre of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran 1417653761, Iran
| | - Dilara Onan
- Department of Physiotherapy and Rehabilitation, Faculty of Heath Sciences, Yozgat Bozok University, Yozgat 66000, Turkey;
| | - Paolo Martelletti
- School of Health, Unitelma Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
2
|
Hoebart C, Kiss A, Podesser BK, Tahir A, Fischer MJM, Heber S. Sensory Neurons Release Cardioprotective Factors in an In Vitro Ischemia Model. Biomedicines 2024; 12:1856. [PMID: 39200320 PMCID: PMC11351881 DOI: 10.3390/biomedicines12081856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Sensory neurons densely innervate the myocardium. The role of their sensing and response to acute and prolonged ischemia is largely unclear. In a cellular model of ischemia-reperfusion injury, the presence of sensory neurons increases cardiomyocyte survival. Here, after the exclusion of classical neurotransmitter release, and measurement of cytokine release, we modified the experiment from a direct co-culture of primary murine cardiomyocytes and sensory neurons to a transfer of the supernatant. Sensory neurons were exposed to ischemia and the resulting conditioned supernatant was transferred onto cardiomyocytes. This approach largely increased the tolerance of cardiomyocytes to ischemia and reperfusion. Towards the identification of the mechanism, it was demonstrated that after ten-fold dilution, the conditioned solution lost its protective effect. The effect remained after removal of extracellular vesicles by ultracentrifugation, and was not affected by exposure to protease activity, and fractionation pointed towards a hydrophilic agent. Solutions conditioned by HEK293t cells or 3T3 fibroblasts also increase cardiomyocyte survival, but to a lower degree. A metabolomic search identified 64 at least two-fold changed metabolites and lipids. Many of these could be identified and are involved in essential cellular functions. In the presented model for ischemia-reperfusion, sensory neurons secrete one or more cardioprotective substances that can improve cardiomyocyte survival.
Collapse
Affiliation(s)
- Clara Hoebart
- Institute of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (C.H.); (S.H.)
| | - Attila Kiss
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria; (A.K.); (B.K.P.)
| | - Bruno K. Podesser
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria; (A.K.); (B.K.P.)
| | - Ammar Tahir
- Division of Pharmacognosy, University of Vienna, 1090 Vienna, Austria;
| | - Michael J. M. Fischer
- Institute of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (C.H.); (S.H.)
| | - Stefan Heber
- Institute of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (C.H.); (S.H.)
| |
Collapse
|
3
|
Tu KY, Jung CJ, Shih YH, Chang ALS. Therapeutic strategies focusing on immune dysregulation and neuroinflammation in rosacea. Front Immunol 2024; 15:1403798. [PMID: 39136023 PMCID: PMC11317294 DOI: 10.3389/fimmu.2024.1403798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
Rosacea is a complex inflammatory condition characterized by papulopustular lesions and erythema on the central face for which there is no cure. The development of rosacea is influenced by both external triggers and genetics, but the common pathophysiology is overactivation of the immune system. Here, we review the current data on proinflammatory cytokines and dysregulation of the neurovascular system as targetable components of rosacea. Amelioration of cutaneous and gastrointestinal dysbiosis and other external factors impacts the immune state and has been observed to improve rosacea. While multiple treatments exist, many patients do not achieve their goals for rosacea control and highlights an unmet need for dermatologic care. Current interventions encompass topical/oral drugs, light devices, and avoidance of triggers management. Additional understanding of the underlying pathogenesis may help us develop novel targeted therapeutic strategies to improve rosacea.
Collapse
Affiliation(s)
- Kuan-Yi Tu
- Division of General Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei, Taiwan
| | - Chiau-Jing Jung
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Hsien Shih
- Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Dermatology, Taipei Medical University Shuang Ho Hospital, New Taipei, Taiwan
| | - Anne Lynn S. Chang
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA, United States
| |
Collapse
|
4
|
Onan D, Farham F, Martelletti P. Clinical Conditions Targeted by OnabotulinumtoxinA in Different Ways in Medicine. Toxins (Basel) 2024; 16:309. [PMID: 39057949 PMCID: PMC11280961 DOI: 10.3390/toxins16070309] [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: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
OnabotulinumtoxinA (BT-A) is used in different medical fields for its beneficial effects. BT-A, a toxin originally produced by the bacterium Clostridium botulinum, is widely known for its ability to temporarily paralyze muscles by blocking the release of acetylcholine, a neurotransmitter involved in muscle contraction. The literature continually reports new hypotheses regarding potential applications that do not consider blockade of acetylcholine release at the neuromuscular junction as a common pathway. In this opinion article, it is our aim to investigate the different pathway targets of BT-A in different medical applications. First of all, the acetylcholine effect of BT-A is used to reduce wrinkles for cosmetic purposes, in the treatment of urological problems, excessive sweating, temporomandibular joint disorders, obesity, migraine, spasticity in neurological diseases, and in various cases of muscle overactivity such as cervical dystonia, blepharospasm, and essential head tremor. In another potential pathway, glutamate A, CGRP, and substance P are targeted for pain inhibition with BT-A application in conditions such as migraine, trigeminal neuralgia, neuropathic pain, and myofascial pain syndrome. On the other hand, as a mechanism different from acetylcholine and pain mediators, BT-A is used in the treatment of hair loss by increasing oxygenation and targeting transforming growth factor-beta 1 cells. In addition, the effect of BT-A on the apoptosis of cancer cells is also known and is being developed. The benefits of BT-A applied in different doses to different regions for different medical purposes are shown in literature studies, and it is also emphasized in those studies that repeating the applications increases the benefits in the long term. The use of BT-A continues to expand as researchers discover new potential therapeutic uses for this versatile toxin.
Collapse
Affiliation(s)
- Dilara Onan
- Department of Physiotherapy and Rehabilitation, Faculty of Heath Sciences, Yozgat Bozok University, Yozgat 66000, Turkey
| | - Fatemeh Farham
- Department of Headache, Iranian Centre of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran 1417653761, Iran;
| | - Paolo Martelletti
- School of Health, Unitelma Sapienza University of Rome, 00161 Rome, Italy;
| |
Collapse
|
5
|
Iadarola MJ, Sapio MR, Loydpierson AJ, Mervis CB, Fehrenbacher JC, Vasko MR, Maric D, Eisenberg DP, Nash TA, Kippenhan JS, Garvey MH, Mannes AJ, Gregory MD, Berman KF. Syntaxin1A overexpression and pain insensitivity in individuals with 7q11.23 duplication syndrome. JCI Insight 2024; 9:e176147. [PMID: 38261410 DOI: 10.1172/jci.insight.176147] [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: 10/18/2023] [Accepted: 01/11/2024] [Indexed: 01/25/2024] Open
Abstract
Genetic modifications leading to pain insensitivity phenotypes, while rare, provide invaluable insights into the molecular biology of pain and reveal targets for analgesic drugs. Pain insensitivity typically results from Mendelian loss-of-function mutations in genes expressed in nociceptive (pain-sensing) dorsal root ganglion (DRG) neurons that connect the body to the spinal cord. We document a pain insensitivity mechanism arising from gene overexpression in individuals with the rare 7q11.23 duplication syndrome (Dup7), who have 3 copies of the approximately 1.5-megabase Williams syndrome (WS) critical region. Based on parental accounts and pain ratings, people with Dup7, mainly children in this study, are pain insensitive following serious injury to skin, bones, teeth, or viscera. In contrast, diploid siblings (2 copies of the WS critical region) and individuals with WS (1 copy) show standard reactions to painful events. A converging series of human assessments and cross-species cell biological and transcriptomic studies identified 1 likely candidate in the WS critical region, STX1A, as underlying the pain insensitivity phenotype. STX1A codes for the synaptic vesicle fusion protein syntaxin1A. Excess syntaxin1A was demonstrated to compromise neuropeptide exocytosis from nociceptive DRG neurons. Taken together, these data indicate a mechanism for producing "genetic analgesia" in Dup7 and offer previously untargeted routes to pain control.
Collapse
Affiliation(s)
- Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Amelia J Loydpierson
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Carolyn B Mervis
- Neurodevelopmental Sciences Laboratory, Department of Psychological and Brain Sciences, University of Louisville, Louisville, Kentucky, USA
| | - Jill C Fehrenbacher
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael R Vasko
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke (NINDS), and
| | - Daniel P Eisenberg
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| | - Tiffany A Nash
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| | - J Shane Kippenhan
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| | - Madeline H Garvey
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Michael D Gregory
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| | - Karen F Berman
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| |
Collapse
|
6
|
Nemanić D, Mustapić M, Matak I, Bach-Rojecky L. Botulinum toxin type a antinociceptive activity in trigeminal regions involves central transcytosis. Eur J Pharmacol 2024; 963:176279. [PMID: 38123005 DOI: 10.1016/j.ejphar.2023.176279] [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: 08/31/2023] [Revised: 11/15/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE Botulinum toxin type A (BoNT-A) provides lasting pain relief in patients with craniofacial pain conditions but the mechanisms of its antinociceptive activity remain unclear. Preclinical research revealed toxin axonal transport to the central afferent terminals, but it is unknown if its central effects involve transsynaptic traffic to the higher-order synapses. To answer this, we examined the contribution of central BoNT-A transcytosis to its action in experimental orofacial pain. MATERIAL AND METHODS Male Wistar rats, 3-4 months old, were injected with BoNT-A (7 U/kg) unilaterally into the vibrissal pad. To investigate the possible contribution of toxin's transcytosis, BoNT-A-neutralizing antiserum (5 IU) was applied intracisternally. Antinocicepive BoNT-A action was assessed by duration of nocifensive behaviors and c-Fos activation in the trigeminal nucleus caudalis (TNC) following bilateral or unilateral formalin (2.5%) application into the vibrissal pad. Additionally, cleaved synaptosomal-associated protein of 25 kDa (cl-SNAP-25) immunoreactivity was analyzed in the bilateral TNC. RESULTS Unilaterally injected BoNT-A reduced the nocifensive behaviors and bilateral c-Fos activation induced by formalin, which was accompanied by the toxin's enzymatic activity on both sides of the TNC. BoNT-A antinociceptive or enzymatic activities were prevented by the specific neutralizing antitoxin. BoNT-A contralateral action occurred independently from ipsilateral side nociception or contralateral trigeminal nerve-mediated axonal traffic. CONCLUSION Herein, we demonstrate that antinociceptive action of pericranially administered BoNT-A involves transsynaptic transport to second order synapses and contralateral trigeminal nociceptive nuclei. These results reveal more complex central toxin activity, necessary to explain its clinical effectiveness in the trigeminal region-related pain states.
Collapse
Affiliation(s)
- Dalia Nemanić
- Department of Pharmacology, University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10 000, Zagreb, Croatia
| | - Matej Mustapić
- Department of Pharmacology, University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10 000, Zagreb, Croatia
| | - Ivica Matak
- Department of Pharmacology, University of Zagreb School of Medicine, Šalata 11, 10 000, Zagreb, Croatia
| | - Lidija Bach-Rojecky
- Department of Pharmacology, University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10 000, Zagreb, Croatia.
| |
Collapse
|
7
|
Yi J, Bertels Z, Del Rosario JS, Widman AJ, Slivicki RA, Payne M, Susser HM, Copits BA, Gereau RW. Bradykinin receptor expression and bradykinin-mediated sensitization of human sensory neurons. Pain 2024; 165:202-215. [PMID: 37703419 PMCID: PMC10723647 DOI: 10.1097/j.pain.0000000000003013] [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: 04/21/2023] [Revised: 06/08/2023] [Accepted: 06/26/2023] [Indexed: 09/15/2023]
Abstract
ABSTRACT Bradykinin is a peptide implicated in inflammatory pain in both humans and rodents. In rodent sensory neurons, activation of B1 and B2 bradykinin receptors induces neuronal hyperexcitability. Recent evidence suggests that human and rodent dorsal root ganglia (DRG), which contain the cell bodies of sensory neurons, differ in the expression and function of key GPCRs and ion channels; whether bradykinin receptor expression and function are conserved across species has not been studied in depth. In this study, we used human DRG tissue from organ donors to provide a detailed characterization of bradykinin receptor expression and bradykinin-induced changes in the excitability of human sensory neurons. We found that B2 and, to a lesser extent, B1 receptors are expressed by human DRG neurons and satellite glial cells. B2 receptors were enriched in the nociceptor subpopulation. Using patch-clamp electrophysiology, we found that acute bradykinin increases the excitability of human sensory neurons, whereas prolonged exposure to bradykinin decreases neuronal excitability in a subpopulation of human DRG neurons. Finally, our analyses suggest that donor's history of chronic pain and age may be predictors of higher B1 receptor expression in human DRG neurons. Together, these results indicate that acute bradykinin-induced hyperexcitability, first identified in rodents, is conserved in humans and provide further evidence supporting bradykinin signaling as a potential therapeutic target for treating pain in humans.
Collapse
Affiliation(s)
- Jiwon Yi
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
- Neuroscience Graduate Program, Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Zachariah Bertels
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - John Smith Del Rosario
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Allie J. Widman
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Richard A. Slivicki
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Maria Payne
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Henry M. Susser
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Bryan A. Copits
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Robert W. Gereau
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
- Department of Neuroscience, Washington University, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
| |
Collapse
|
8
|
Montalbetti N, Dalghi MG, Parakala-Jain T, Clayton D, Apodaca G, Carattino MD. Antinociceptive effect of the calcitonin gene-related peptide receptor antagonist BIBN4096BS in mice with bacterial cystitis. Am J Physiol Renal Physiol 2023; 325:F779-F791. [PMID: 37823199 PMCID: PMC10878727 DOI: 10.1152/ajprenal.00217.2023] [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: 07/25/2023] [Revised: 09/25/2023] [Accepted: 10/09/2023] [Indexed: 10/13/2023] Open
Abstract
Patients with urinary tract infections (UTIs) suffer from urinary frequency, urgency, dysuria, and suprapubic pain, but the mechanisms by which bladder afferents sense the presence of uropathogens and encode this information is not well understood. Calcitonin gene-related peptide (CGRP) is a 37-mer neuropeptide found in a subset of bladder afferents that terminate primarily in the lamina propria. Here, we report that the CGRP receptor antagonist BIBN4096BS lessens lower urinary tract symptoms and prevents the development of pelvic allodynia in mice inoculated with uropathogenic Escherichia coli (UPEC) without altering urine bacterial loads or the host immune response to the infection. These findings indicate that CGRP facilitates the processing of noxious/inflammatory stimuli during UPEC infection. Using fluorescent in situ hybridization, we identified a population of suburothelial fibroblasts in the lamina propria, a region where afferent fibers containing CGRP terminate, that expresses the canonical CGRP receptor components Calcrl and Ramp1. We propose that these fibroblasts, in conjunction with CGRP+ afferents, form a circuit that senses substances released during the infection and transmit this noxious information to the central nervous system.NEW & NOTEWORTHY Afferent C fibers release neuropeptides including calcitonin gene-related peptide (CGRP). Here, we show that the specific CGRP receptor antagonist, BIBN409BS, ameliorates lower urinary tract symptoms and pelvic allodynia in mice inoculated with uropathogenic E. coli. Using fluorescent in situ hybridization, we identified a population of suburothelial fibroblasts in the lamina propria that expresses the canonical CGRP receptor. Our findings indicate that CGRP contributes to the transmission of nociceptive information arising from the bladder.
Collapse
Affiliation(s)
- Nicolas Montalbetti
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Marianela G Dalghi
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Tanmay Parakala-Jain
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Dennis Clayton
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Gerard Apodaca
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Marcelo D Carattino
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
9
|
Dai X, Li L, Yan X, Fan Q, Wang R, Zhang W, Chen W, Liu Y, Meng J, Wang J. Myeloid Vamp3 deletion attenuates CFA-induced inflammation and pain in mice via ameliorating macrophage infiltration and inflammatory cytokine production. Front Immunol 2023; 14:1239592. [PMID: 37965323 PMCID: PMC10641732 DOI: 10.3389/fimmu.2023.1239592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
Persistent inflammation and associated pain significantly impact individuals' quality of life, posing substantial healthcare challenges. Proinflammatory cytokines, released by activated macrophages, play crucial roles in the development of chronic inflammatory conditions such as rheumatoid arthritis. To identify and evaluate potential therapeutic interventions targeting this process for mitigating inflammation and pain, we created myeloid cell-specific knockout of Vamp3 (vesicle-associated membrane protein 3) mice (Vamp3 Δmyel) by crossing LysM-Cre mice with newly engineered Vamp3flox/flox mice. Bone marrow-derived macrophages and peritoneal resident macrophages from Vamp3 Δmyel mice exhibited a significant reduction in TNF-α and IL-6 release compared to control mice. Moreover, Vamp3 deficiency led to decreased paw edema and ankle joint swelling induced by intraplantar injection of complete Freund's adjuvant (CFA). Furthermore, Vamp3 depletion also mitigated CFA-induced mechanical allodynia and thermal hyperalgesia. Mechanistically, Vamp3 loss ameliorated the infiltration of macrophages in peripheral sites of the hind paw and resulted in reduced levels of TNF-α and IL-6 in the CFA-injected paw and serum. RT-qPCR analysis demonstrated downregulation of various inflammation-associated genes, including TNF-α, IL-6, IL-1β, CXCL11, TIMP-1, COX-2, CD68, and CD54 in the injected paw at the test day 14 following CFA administration. These findings highlight the novel role of Vamp3 in regulating inflammatory responses and suggest it as a potential therapeutic target for the development of novel Vamp-inactivating therapeutics, with potential applications in the management of inflammatory diseases.
Collapse
Affiliation(s)
- Xiaolong Dai
- School of Life Sciences, Henan University, Kaifeng, China
| | - Lianlian Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Xinrong Yan
- School of Life Sciences, Henan University, Kaifeng, China
| | - Qianqian Fan
- School of Life Sciences, Henan University, Kaifeng, China
| | - Ruizhen Wang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Wenhao Zhang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Weiwei Chen
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yang Liu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jianghui Meng
- School of Life Sciences, Henan University, Kaifeng, China
- School of Biotechnology, Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Jiafu Wang
- School of Life Sciences, Henan University, Kaifeng, China
- School of Biotechnology, Faculty of Science and Health, Dublin City University, Dublin, Ireland
| |
Collapse
|
10
|
Bonura A, Brunelli N, Marcosano M, Iaccarino G, Fofi L, Vernieri F, Altamura C. Calcitonin Gene-Related Peptide Systemic Effects: Embracing the Complexity of Its Biological Roles-A Narrative Review. Int J Mol Sci 2023; 24:13979. [PMID: 37762283 PMCID: PMC10530509 DOI: 10.3390/ijms241813979] [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: 07/28/2023] [Revised: 08/30/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The calcitonin gene-related peptide (CGRP) is a neuropeptide widely distributed throughout the human body. While primarily recognized as a nociceptive mediator, CGRP antagonists are currently utilized for migraine treatment. However, its role extends far beyond this, acting as a regulator of numerous biological processes. Indeed, CGRP plays a crucial role in vasodilation, inflammation, intestinal motility, and apoptosis. In this review, we explore the non-nociceptive effects of CGRP in various body systems, revealing actions that can be contradictory at times. In the cardiovascular system, it functions as a potent vasodilator, yet its antagonists do not induce arterial hypertension, suggesting concurrent modulation by other molecules. As an immunomodulator, CGRP exhibits intriguing complexity, displaying both anti-inflammatory and pro-inflammatory effects. Furthermore, CGRP appears to be involved in obesity development while paradoxically reducing appetite. A thorough investigation of CGRP's biological effects is crucial for anticipating potential side effects associated with its antagonists' use and for developing novel therapies in other medical fields. In summary, CGRP represents a neuropeptide with a complex systemic impact, extending well beyond nociception, thus offering new perspectives in medical research and therapeutics.
Collapse
Affiliation(s)
- Adriano Bonura
- Instituite of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (A.B.); (N.B.); (M.M.); (L.F.); (F.V.)
- Unit of Headache and Neurosonology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Nicoletta Brunelli
- Instituite of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (A.B.); (N.B.); (M.M.); (L.F.); (F.V.)
- Unit of Headache and Neurosonology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Marilena Marcosano
- Instituite of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (A.B.); (N.B.); (M.M.); (L.F.); (F.V.)
- Unit of Headache and Neurosonology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Gianmarco Iaccarino
- Instituite of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (A.B.); (N.B.); (M.M.); (L.F.); (F.V.)
- Unit of Headache and Neurosonology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Luisa Fofi
- Instituite of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (A.B.); (N.B.); (M.M.); (L.F.); (F.V.)
- Unit of Headache and Neurosonology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Fabrizio Vernieri
- Instituite of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (A.B.); (N.B.); (M.M.); (L.F.); (F.V.)
- Unit of Headache and Neurosonology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Claudia Altamura
- Instituite of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (A.B.); (N.B.); (M.M.); (L.F.); (F.V.)
- Unit of Headache and Neurosonology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| |
Collapse
|
11
|
Chen C, Wang P, Zhang L, Liu X, Zhang H, Cao Y, Wang X, Zeng Q. Exploring the Pathogenesis and Mechanism-Targeted Treatments of Rosacea: Previous Understanding and Updates. Biomedicines 2023; 11:2153. [PMID: 37626650 PMCID: PMC10452301 DOI: 10.3390/biomedicines11082153] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Rosacea is a chronic inflammatory skin disease characterized by recurrent erythema, flushing, telangiectasia, papules, pustules, and phymatous changes in the central area of the face. Patients with this condition often experience a significant negative impact on their quality of life, self-esteem, and overall well-being. Despite its prevalence, the pathogenesis of rosacea is not yet fully understood. Recent research advances are reshaping our understanding of the underlying mechanisms of rosacea, and treatment options based on the pathophysiological perspective hold promise to improve patient outcomes and reduce incidence. In this comprehensive review, we investigate the pathogenesis of rosacea in depth, with a focus on emerging and novel mechanisms, and provide an up-to-date overview of therapeutic strategies that target the diverse pathogenic mechanisms of rosacea. Lastly, we discuss potential future research directions aimed at enhancing our understanding of the condition and developing effective treatments.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiuli Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200040, China
| | - Qingyu Zeng
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200040, China
| |
Collapse
|
12
|
Brin MF, Blitzer A. The pluripotential evolution and journey of Botox (onabotulinumtoxinA). Medicine (Baltimore) 2023; 102:e32373. [PMID: 37499079 PMCID: PMC10374190 DOI: 10.1097/md.0000000000032373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
Clinical use of onabotulinumtoxinA evolved based on strategic, hypothesis-driven applications, as well as serendipitous observations by physicians and patients. The success of onabotulinumtoxinA in blepharospasm and strabismus led to its study in other head and neck dystonias, followed by limb dystonia, tremor, and spasticity. The aesthetic use of onabotulinumtoxinA followed initial reports from patients of improved facial lines after injections for facial dystonias and hemifacial spasm. Although patients with dystonias and spasticity regularly reported that their local pain improved after injections, onabotulinumtoxinA was not systematically explored for chronic migraine until patients began reporting headache improvements following aesthetic injections. Clinicians began assessing onabotulinumtoxinA for facial sweating and hyperhidrosis based on its inhibition of acetylcholine from sympathetic cholinergic nerves. Yet another line of research grew out of injections for laryngeal dystonia, whereby clinicians began to explore other sphincters in the gastrointestinal tract and eventually to treatment of pelvic sphincters; many of these sphincters are innervated by autonomic nerves. Additional investigations in other autonomically mediated conditions were conducted, including overactive bladder and neurogenic detrusor overactivity, achalasia, obesity, and postoperative atrial fibrillation. The study of onabotulinumtoxinA for depression also grew out of the cosmetic experience and the observation that relaxing facial muscle contractions associated with negative emotions may improve mood. For approved indications, the safety profile of onabotulinumtoxinA has been demonstrated in the formal development programs and post-marketing reports. Over time, evidence has accumulated suggesting clinical manifestations of systemic effects, albeit uncommon, particularly with high doses and in vulnerable populations. Although onabotulinumtoxinA is approved for approximately 26 indications across multiple local regions, there are 15 primary indication uses that have been approved in most regions, including the United States, Europe, South America, and Asia. This review describes many uses for which AbbVie has not sought and/or received regulatory approval and are mentioned for historical context only.
Collapse
Affiliation(s)
- Mitchell F Brin
- Allergan/AbbVie, Irvine, CA, USA
- University of California, Irvine, CA, USA
| | - Andrew Blitzer
- Professor Emeritus of Otolaryngology/Head and Neck Surgery, Columbia University College of Physicians and Surgeons, New York, NY, USA
- Adjunct Professor of Neurology, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Director, NY Center for Voice and Swallowing Disorders, New York, NY, USA
- Co-founder and Director of Research, ADN International, New York, NY, USA
| |
Collapse
|
13
|
Huang LS, Anas M, Xu J, Zhou B, Toth PT, Krishnan Y, Di A, Malik AB. Endosomal trafficking of two-pore K + efflux channel TWIK2 to plasmalemma mediates NLRP3 inflammasome activation and inflammatory injury. eLife 2023; 12:e83842. [PMID: 37158595 PMCID: PMC10202452 DOI: 10.7554/elife.83842] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 05/07/2023] [Indexed: 05/10/2023] Open
Abstract
Potassium efflux via the two-pore K+ channel TWIK2 is a requisite step for the activation of NLRP3 inflammasome, however, it remains unclear how K+ efflux is activated in response to select cues. Here, we report that during homeostasis, TWIK2 resides in endosomal compartments. TWIK2 is transported by endosomal fusion to the plasmalemma in response to increased extracellular ATP resulting in the extrusion of K+. We showed that ATP-induced endosomal TWIK2 plasmalemma translocation is regulated by Rab11a. Deleting Rab11a or ATP-ligated purinergic receptor P2X7 each prevented endosomal fusion with the plasmalemma and K+ efflux as well as NLRP3 inflammasome activation in macrophages. Adoptive transfer of Rab11a-depleted macrophages into mouse lungs prevented NLRP3 inflammasome activation and inflammatory lung injury. We conclude that Rab11a-mediated endosomal trafficking in macrophages thus regulates TWIK2 localization and activity at the cell surface and the downstream activation of the NLRP3 inflammasome. Results show that endosomal trafficking of TWIK2 to the plasmalemma is a potential therapeutic target in acute or chronic inflammatory states.
Collapse
Affiliation(s)
- Long Shuang Huang
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong UniversityShanghaiChina
| | - Mohammad Anas
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
| | - Jingsong Xu
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
| | - Bisheng Zhou
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
| | - Peter T Toth
- Fluorescence Imaging Core, The University of Illinois College of MedicineChicagoUnited States
| | - Yamuna Krishnan
- Department of Chemistry, University of ChicagoChicagoUnited States
| | - Anke Di
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
| | - Asrar B Malik
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of MedicineChicagoUnited States
| |
Collapse
|
14
|
Yi J, Bertels Z, Del Rosario JS, Widman AJ, Slivicki RA, Payne M, Susser HM, Copits BA, Gereau RW. Bradykinin receptor expression and bradykinin-mediated sensitization of human sensory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.31.534820. [PMID: 37034782 PMCID: PMC10081334 DOI: 10.1101/2023.03.31.534820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Bradykinin is a peptide implicated in inflammatory pain in both humans and rodents. In rodent sensory neurons, activation of B1 and B2 bradykinin receptors induces neuronal hyperexcitability. Recent evidence suggests that human and rodent dorsal root ganglia (DRG), which contain the cell bodies of sensory neurons, differ in the expression and function of key GPCRs and ion channels; whether BK receptor expression and function are conserved across species has not been studied in depth. In this study, we used human DRG tissue from organ donors to provide a detailed characterization of bradykinin receptor expression and bradykinin-induced changes in the excitability of human sensory neurons. We found that B2 and, to a lesser extent, B1 receptors are expressed by human DRG neurons and satellite glial cells. B2 receptors were enriched in the nociceptor subpopulation. Using patch-clamp electrophysiology, we found that acute bradykinin increases the excitability of human sensory neurons, while prolonged exposure to bradykinin decreases neuronal excitability in a subpopulation of human DRG neurons. Finally, our analyses suggest that donor’s history of chronic pain and age may be predictors of higher B1 receptor expression in human DRG neurons. Together, these results indicate that acute BK-induced hyperexcitability, first identified in rodents, is conserved in humans and provide further evidence supporting BK signaling as a potential therapeutic target for treating pain in humans.
Collapse
Affiliation(s)
- Jiwon Yi
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Zachariah Bertels
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - John Smith Del Rosario
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Allie J. Widman
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Richard A. Slivicki
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Maria Payne
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Henry M. Susser
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Bryan A. Copits
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Robert W. Gereau
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
- Department of Neuroscience, Washington University, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
| |
Collapse
|
15
|
Sudershan A, Younis M, Sudershan S, Kumar P. Migraine as an inflammatory disorder with microglial activation as a prime candidate. Neurol Res 2023; 45:200-215. [PMID: 36197286 DOI: 10.1080/01616412.2022.2129774] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2022]
Abstract
BACKGROUND The lower threshold of neuronal hyperexcitability has been correlated with migraines for decades but as technology has progressed, it has now become conceivable to learn more about the migraine disease. Apart from the "cortical spreading depression" and "activation of the trigeminovascular system", inflammation has been increasingly recognized as a possible pathogenic process that may have the possibility to regulate the disease severity. Microglial cells, the prime candidate of the innate immune cells of central nervous tissue, has been associated with numerous diseases; including cancer, neurodegenerative disorders, and inflammatory disorders. AIM In this review, we have attempted to link the dot of various microglial activation signaling pathways to enlighten the correlation between microglial involvement and the progression of migraine conditions. METHOD A structured survey of research articles and review of the literature was done in the electronic databases of Google Scholar, PubMed, Springer, and Elsevier until 31 December 2021. RESULT & CONCLUSION Of 1136 articles found initially and screening of 1047 records, 47 studies were included for the final review. This review concluded that inflammation and microglial overexpression as the prime candidate, plays an important role in the modulation of migraine and are responsible for the progression toward chronification. Therefore, this increases the possibility of preventing migraine development and chronification by blocking microglia overexpression.
Collapse
Affiliation(s)
- Amrit Sudershan
- Institute of Human Genetics, University of Jammu, Jammu and Kashmir 180006, India
| | - Mohd Younis
- Department of Human Genetics and Molecular Biology, Bharathair University, Coimbatore, 641046, India
| | - Srishty Sudershan
- Department of Zoology, University of Jammu, Jammu and Kashmir, 180006, India
| | - Parvinder Kumar
- Institute of Human Genetics, University of Jammu, Jammu and Kashmir 180006, India.,Department of Zoology, University of Jammu, Jammu and Kashmir, 180006, India
| |
Collapse
|
16
|
Moore AA, Nelson M, Wickware C, Choi S, Moon G, Xiong E, Orta L, Brideau-Andersen A, Brin MF, Broide RS, Liedtke W, Moore C. OnabotulinumtoxinA effects on trigeminal nociceptors. Cephalalgia 2023; 43:3331024221141683. [PMID: 36751871 PMCID: PMC10652784 DOI: 10.1177/03331024221141683] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
BACKGROUND OnabotulinumtoxinA (onabotA) is approved globally for prevention of chronic migraine; however, the classical mechanism of action of onabotA in motor and autonomic neurons cannot fully explain the effectiveness of onabotulinumtoxinA in this sensory neurological disease. We sought to explore the direct effects of onabotulinumtoxinA on mouse trigeminal ganglion sensory neurons using an inflammatory soup-based model of sensitization. METHODS Primary cultured trigeminal ganglion neurons were pre-treated with inflammatory soup, then treated with onabotulinumtoxinA (2.75 pM). Treated neurons were used to examine transient receptor potential vanilloid subtype 1 and transient receptor potential ankyrin 1 cell-surface expression, calcium influx, and neuropeptide release. RESULTS We found that onabotulinumtoxinA cleaved synaptosomal-associated protein-25 kDa in cultured trigeminal ganglion neurons; synaptosomal-associated protein-25 kDa cleavage was enhanced by inflammatory soup pre-treatment, suggesting greater uptake of toxin under sensitized conditions. OnabotulinumtoxinA also prevented inflammatory soup-mediated increases in TRPV1 and TRPA1 cell-surface expression, without significantly altering TRPV1 or TRPA1 protein expression in unsensitized conditions. We observed similar inhibitory effects of onabotulinumtoxinA on TRP-mediated calcium influx and TRPV1- and TRPA1-mediated release of calcitonin gene-related peptide and prostaglandin 2 under sensitized, but not unsensitized control, conditions. CONCLUSIONS Our data deepen the understanding of the sensory mechanism of action of onabotulinumtoxinA and support the notion that, once endocytosed, the cytosolic light chain of onabotulinumtoxinA cleaves synaptosomal-associated protein-25 kDa to prevent soluble N-ethylmaleimide-sensitive factor attachment protein receptor-mediated processes more generally in motor, autonomic, and sensory neurons.
Collapse
Affiliation(s)
- Ashley A Moore
- Department of Neurology, Duke University, Durham, NC, USA
| | | | | | - Shinbe Choi
- Department of Neurology, Duke University, Durham, NC, USA
| | - Gene Moon
- Department of Neurology, Duke University, Durham, NC, USA
| | - Emma Xiong
- Department of Neurology, Duke University, Durham, NC, USA
| | - Lily Orta
- Department of Neurology, Duke University, Durham, NC, USA
| | | | - Mitchell F Brin
- Allergan, an AbbVie company, Irvine, CA, USA
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | | | - Wolfgang Liedtke
- Department of Neurology, Duke University, Durham, NC, USA
- Department of Molecular Pathobiology – Dental Pain Research, New York University College of Dentistry, New York, NY, USA
| | - Carlene Moore
- Department of Neurology, Duke University, Durham, NC, USA
| |
Collapse
|
17
|
Belinskaia M, Wang J, Kaza SK, Antoniazzi C, Zurawski T, Dolly JO, Lawrence GW. Bipartite Activation of Sensory Neurons by a TRPA1 Agonist Allyl Isothiocyanate Is Reflected by Complex Ca 2+ Influx and CGRP Release Patterns: Enhancement by NGF and Inhibition with VAMP and SNAP-25 Cleaving Botulinum Neurotoxins. Int J Mol Sci 2023; 24:ijms24021338. [PMID: 36674850 PMCID: PMC9865456 DOI: 10.3390/ijms24021338] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
The trafficking of transient receptor potential (TRP) channels to the plasma membrane and the release of calcitonin gene-related peptide (CGRP) from trigeminal ganglion neurons (TGNs) are implicated in some aspects of chronic migraines. These exocytotic processes are inhibited by cleavage of SNAREs with botulinum neurotoxins (BoNTs); moreover, type A toxin (/A) clinically reduces the frequency and severity of migraine attacks but not in all patients for unknown reasons. Herein, neonatal rat TGNs were stimulated with allyl isothiocyanate (AITC), a TRPA1 agonist, and dose relationships were established to link the resultant exocytosis of CGRP with Ca2+ influx. The CGRP release, quantified by ELISA, was best fit by a two-site model (EC50 of 6 and 93 µM) that correlates with elevations in intracellular Ca2+ [Ca2+]i revealed by time-lapse confocal microscopy of fluo-4-acetoxymethyl ester (Fluo-4 AM) loaded cells. These signals were all blocked by two TRPA1 antagonists, HC-030031 and A967079. At low [AITC], [Ca2+]i was limited because of desensitisation to the agonist but rose for concentrations > 0.1 mM due to a deduced non-desensitising second phase of Ca2+ influx. A recombinant BoNT chimera (/DA), which cleaves VAMP1/2/3, inhibited AITC-elicited CGRP release to a greater extent than SNAP-25-cleaving BoNT/A. /DA also proved more efficacious against CGRP efflux evoked by a TRPV1 agonist, capsaicin. Nerve growth factor (NGF), a pain-inducing sensitiser of TGNs, enhanced the CGRP exocytosis induced by low [AITC] only. Both toxins blocked NGF-induced neuropeptide secretion and its enhancement of the response to AITC. In conclusion, NGF sensitisation of sensory neurons involves TRPA1, elevated Ca2+ influx, and CGRP exocytosis, mediated by VAMP1/2/3 and SNAP-25 which can be attenuated by the BoNTs.
Collapse
|
18
|
Onan D, Bentivegna E, Martelletti P. OnabotulinumtoxinA Treatment in Chronic Migraine: Investigation of Its Effects on Disability, Headache and Neck Pain Intensity. Toxins (Basel) 2022; 15:29. [PMID: 36668849 PMCID: PMC9862733 DOI: 10.3390/toxins15010029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/12/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Neck disability and pain are frequently encountered problems in patients with chronic migraine (CM). The long-term stimuli of neurons in the trigeminocervical junction may explain this situation. OnabotulinumtoxinA (ONA) treatment is one of the proven treatments for CM; however, there is no study data on the efficacy of ONA treatment on neck disability and pain in CM patients. Therefore, we aimed to investigate the effect of ONA treatment on disability, neck pain and headache intensity in CM patients. One hundred thirty-four patients who met the inclusion criteria were included in the study. ONA treatment was administered at a dose of 195 U to 39 sites in total as per Follow-the-Pain PREEMPT protocol. The disability was evaluated with the Neck Disability Index and the Migraine Disability Assessment; pain intensity was evaluated with the Visual Analogue Scale; the monthly number of headache days were recorded; quality of life was evaluated with the Headache Impact Test. All assessments were recorded at baseline and 3 months after treatment. After the treatment, neck−migraine disabilities decreased from severe to mild for neck and moderate for migraine (p < 0.001). Neck pain and headache intensities decreased by almost half (p < 0.001). The median number of monthly headache days decreased from 20 days to 6 days (p < 0.000). The quality-of-life level decreased significantly from severe to substantial level (p < 0.001). According to our results, ONA treatment was effective in reducing neck-related problems in CM patients. Long-term follow-up results may provide researchers with more comprehensive results in terms of the treatment of chronic migraine−neck-related problems.
Collapse
Affiliation(s)
- Dilara Onan
- Spine Health Unit, Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara 06230, Türkiye
- Department of Clinical and Molecular Medicine, Sapienza University, 000189 Rome, Italy
| | - Enrico Bentivegna
- Department of Clinical and Molecular Medicine, Sapienza University, 000189 Rome, Italy
| | - Paolo Martelletti
- Department of Clinical and Molecular Medicine, Sapienza University, 000189 Rome, Italy
- Regional Referral Headache Centre, Sant’Andrea Hospital, 000189 Rome, Italy
| |
Collapse
|
19
|
Natural Herbal Non-Opioid Topical Pain Relievers-Comparison with Traditional Therapy. Pharmaceutics 2022; 14:pharmaceutics14122648. [PMID: 36559142 PMCID: PMC9785912 DOI: 10.3390/pharmaceutics14122648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 12/04/2022] Open
Abstract
Pain is the predominant symptom of many clinical diseases and is frequently associated with neurological and musculoskeletal problems. Chronic pain is frequent in the elderly, causing suffering, disability, social isolation, and increased healthcare expenses. Chronic pain medication is often ineffective and has many side effects. Nonsteroidal over-the-counter and prescription drugs are frequently recommended as first-line therapies for pain control; however, long-term safety issues must not be neglected. Herbs and nutritional supplements may be a safer and more effective alternative to nonsteroidal pharmaceuticals for pain management, especially when used long-term. Recently, topical analgesic therapies have gained attention as an innovative approach due to their sufficient efficacy and comparatively fewer systemic side effects and drug-drug interactions. In this paper, we overview the main natural herbal pain relievers, their efficacy and safety, and their potential use as topical agents for pain control. Although herbal-derived medications are not appropriate for providing quick relief for acute pain problems, they could be used as potent alternative remedies in managing chronic persistent pain with minimal side effects.
Collapse
|
20
|
Puri S, Kenyon BM, Hamrah P. Immunomodulatory Role of Neuropeptides in the Cornea. Biomedicines 2022; 10:1985. [PMID: 36009532 PMCID: PMC9406019 DOI: 10.3390/biomedicines10081985] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/21/2022] Open
Abstract
The transparency of the cornea along with its dense sensory innervation and resident leukocyte populations make it an ideal tissue to study interactions between the nervous and immune systems. The cornea is the most densely innervated tissue of the body and possesses both immune and vascular privilege, in part due to its unique repertoire of resident immune cells. Corneal nerves produce various neuropeptides that have a wide range of functions on immune cells. As research in this area expands, further insights are made into the role of neuropeptides and their immunomodulatory functions in the healthy and diseased cornea. Much remains to be known regarding the details of neuropeptide signaling and how it contributes to pathophysiology, which is likely due to complex interactions among neuropeptides, receptor isoform-specific signaling events, and the inflammatory microenvironment in disease. However, progress in this area has led to an increase in studies that have begun modulating neuropeptide activity for the treatment of corneal diseases with promising results, necessitating the need for a comprehensive review of the literature. This review focuses on the role of neuropeptides in maintaining the homeostasis of the ocular surface, alterations in disease settings, and the possible therapeutic potential of targeting these systems.
Collapse
Affiliation(s)
- Sudan Puri
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Brendan M. Kenyon
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Pedram Hamrah
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
- Departments of Immunology and Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
- Cornea Service, Tufts New England Eye Center, Boston, MA 02111, USA
| |
Collapse
|
21
|
Botulinum Toxin Use for Modulating Neuroimmune Cutaneous Activity in Psoriasis. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58060813. [PMID: 35744076 PMCID: PMC9228985 DOI: 10.3390/medicina58060813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022]
Abstract
Psoriasis is a complex immune-mediated inflammatory disorder that generates enormous interest within the scientific communities worldwide, with new therapeutic targets being constantly identified and tested. Despite the numerous topical and systemic medications available for the treatment of psoriasis, alternative therapies are still needed for the optimal management of some patients who present with localized, resistant lesions. Novel insights into the contribution of cutaneous neurogenic inflammation in the pathogenesis of psoriasis have yielded exciting new potential roles of nerve-targeting treatments, namely botulinum toxin type A (BoNT-A), for the management of this disease. This paper aims to review the existing literature on knowledge regarding the potential role of BoNT-A in psoriasis treatment, with a focus on its ability to interfere with the immunopathogenetic aspects of psoriatic disease. Furthermore, in our paper, we are also including the first report of psoriatic lesions remission following local BoNT-A injections that were administered for treating upper limb spasticity, in a patient that concomitantly suffered from psoriasis and post-stroke spasticity.
Collapse
|
22
|
Hary V, Schitter S, Martinez V. Efficacy and safety of botulinum A toxin for the treatment of chronic peripheral neuropathic pain: A systematic review of randomized controlled trials and meta-analysis. Eur J Pain 2022; 26:980-990. [PMID: 35293078 DOI: 10.1002/ejp.1941] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND OBJECTIVE Botulinum toxin type A (BTX-A) is a recently developed treatment for the management of peripheral neuropathic pain. The objective of this study was to provide a synthesis of the evidence concerning the efficacy and safety of subcutaneous botulinum toxin type A injections. DATABASES AND DATA TREATMENT We searched the MEDLINE, EMBASE, LILACS, Cochrane, and Clinical Trial Register databases for randomized controlled trials comparing subcutaneous BTX-A to placebo injections for treating chronic peripheral neuropathic pain. The primary endpoint was the assessment of pain 1 month after the injection. The secondary outcomes were the assessment of pain at 3 months, neuropathic pain intensity and quality of life at 1 and 3 months, and adverse effects. A random-effect meta-analysis was performed on the combined data. Evidence quality was rated by the Grades of Recommendation, Assessment, Development, and Evaluation (GRADE) method. RESULTS Ten randomized controlled trials including 505 patients were included in this review (registration number CRD42021239108). At 1 and 3 months after injection, the BTX-A groups had a lower mean difference (MD) in pain score (MD -1.87 (confidence intervals [CIs] -2.91; -0.83) and -1.38 (CI -1.95; -0.81), respectively). Subgroup analysis showed greater efficacy for diabetic polyneuropathy (MD -2.48, [-3.22; -1.74]). We found no impact of BTX-A on quality of life and no difference in adverse effect between BTX-A and placebo. The evidence was considered of moderate quality. CONCLUSION The pooled data suggest that subcutaneous BTX-A injections have a clinically significant effect, decreasing pain for three months after the injection, but no benefit in terms of quality of life has yet been demonstrated. SIGNIFICANCE We found that botulinum toxin is efficient and safe for the treatment of neuropathic pain, especially for diabetic polyneuropathy. Botulinum toxin type A, used for years in neurology, rehabilitation and physical medicine, has proved innocuous and effective, and should be considered as a serious alternative for pain treatment.
Collapse
Affiliation(s)
- Vincent Hary
- Faculty of medecine, Université de Paris, Paris, France
- Pain Unit, Hôpital Raymond Poincaré, APHP, Garches, France
| | - Sebastien Schitter
- Pain Unit, Hôpital Raymond Poincaré, APHP, Garches, France
- Faculty de medecine, Université Versailles Saint Quentin, Versailles, France
| | - Valeria Martinez
- Pain Unit, Hôpital Raymond Poincaré, APHP, Garches, France
- Faculty de medecine, Université Versailles Saint Quentin, Versailles, France
- INSERM U 987, CETD, Hôpital Ambroise Paré, APHP, Boulogne-Billancourt, France
- Department of Anesthesiology, Hôpital Raymond Poincaré, APHP, Garches, France
| |
Collapse
|
23
|
Antoniazzi C, Belinskaia M, Zurawski T, Kaza SK, Dolly JO, Lawrence GW. Botulinum Neurotoxin Chimeras Suppress Stimulation by Capsaicin of Rat Trigeminal Sensory Neurons In Vivo and In Vitro. Toxins (Basel) 2022; 14:116. [PMID: 35202143 PMCID: PMC8878885 DOI: 10.3390/toxins14020116] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/14/2022] [Accepted: 02/01/2022] [Indexed: 02/06/2023] Open
Abstract
Chimeras of botulinum neurotoxin (BoNT) serotype A (/A) combined with /E protease might possess improved analgesic properties relative to either parent, due to inheriting the sensory neurotropism of the former with more extensive disabling of SNAP-25 from the latter. Hence, fusions of /E protease light chain (LC) to whole BoNT/A (LC/E-BoNT/A), and of the LC plus translocation domain (HN) of /E with the neuronal acceptor binding moiety (HC) of /A (BoNT/EA), created previously by gene recombination and expression in E. coli., were used. LC/E-BoNT/A (75 units/kg) injected into the whisker pad of rats seemed devoid of systemic toxicity, as reflected by an absence of weight loss, but inhibited the nocifensive behavior (grooming, freezing, and reduced mobility) induced by activating TRPV1 with capsaicin, injected at various days thereafter. No sex-related differences were observed. c-Fos expression was increased five-fold in the trigeminal nucleus caudalis ipsi-lateral to capsaicin injection, relative to the contra-lateral side and vehicle-treated controls, and this increase was virtually prevented by LC/E-BoNT/A. In vitro, LC/E-BoNT/A or /EA diminished CGRP exocytosis from rat neonate trigeminal ganglionic neurons stimulated with up to 1 µM capsaicin, whereas BoNT/A only substantially reduced the release in response to 0.1 µM or less of the stimulant, in accordance with the /E protease being known to prevent fusion of exocytotic vesicles.
Collapse
Affiliation(s)
| | | | | | | | | | - Gary W. Lawrence
- International Centre for Neurotherapeutics, Dublin City University, Collins Avenue, D09 V209 Dublin, Ireland; (C.A.); (M.B.); (T.Z.); (S.K.K.); (J.O.D.)
| |
Collapse
|
24
|
Belinskaia M, Zurawski T, Kaza SK, Antoniazzi C, Dolly JO, Lawrence GW. NGF Enhances CGRP Release Evoked by Capsaicin from Rat Trigeminal Neurons: Differential Inhibition by SNAP-25-Cleaving Proteases. Int J Mol Sci 2022; 23:ijms23020892. [PMID: 35055082 PMCID: PMC8778182 DOI: 10.3390/ijms23020892] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/08/2022] [Accepted: 01/11/2022] [Indexed: 11/23/2022] Open
Abstract
Nerve growth factor (NGF) is known to intensify pain in various ways, so perturbing pertinent effects without negating its essential influences on neuronal functions could help the search for much-needed analgesics. Towards this goal, cultured neurons from neonatal rat trigeminal ganglia—a locus for craniofacial sensory nerves—were used to examine how NGF affects the Ca2+-dependent release of a pain mediator, calcitonin gene-related peptide (CGRP), that is triggered by activating a key signal transducer, transient receptor potential vanilloid 1 (TRPV1) with capsaicin (CAP). Measurements utilised neurons fed with or deprived of NGF for 2 days. Acute re-introduction of NGF induced Ca2+-dependent CGRP exocytosis that was inhibited by botulinum neurotoxin type A (BoNT/A) or a chimera of/E and/A (/EA), which truncated SNAP-25 (synaptosomal-associated protein with Mr = 25 k) at distinct sites. NGF additionally caused a Ca2+-independent enhancement of the neuropeptide release evoked by low concentrations (<100 nM) of CAP, but only marginally increased the peak response to ≥100 nM. Notably, BoNT/A inhibited CGRP exocytosis evoked by low but not high CAP concentrations, whereas/EA effectively reduced responses up to 1 µM CAP and inhibited to a greater extent its enhancement by NGF. In addition to establishing that sensitisation of sensory neurons to CAP by NGF is dependent on SNARE-mediated membrane fusion, insights were gleaned into the differential ability of two regions in the C-terminus of SNAP-25 (181–197 and 198–206) to support CAP-evoked Ca2+-dependent exocytosis at different intensities of stimulation.
Collapse
|
25
|
Mi J, Xu J, Yao Z, Yao H, Li Y, He X, Dai B, Zou L, Tong W, Zhang X, Hu P, Ruan YC, Tang N, Guo X, Zhao J, He J, Qin L. Implantable Electrical Stimulation at Dorsal Root Ganglions Accelerates Osteoporotic Fracture Healing via Calcitonin Gene-Related Peptide. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103005. [PMID: 34708571 PMCID: PMC8728818 DOI: 10.1002/advs.202103005] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/11/2021] [Indexed: 05/18/2023]
Abstract
The neuronal engagement of the peripheral nerve system plays a crucial role in regulating fracture healing, but how to modulate the neuronal activity to enhance fracture healing remains unexploited. Here it is shown that electrical stimulation (ES) directly promotes the biosynthesis and release of calcitonin gene-related peptide (CGRP) by activating Ca2+ /CaMKII/CREB signaling pathway and action potential, respectively. To accelerate rat femoral osteoporotic fracture healing which presents with decline of CGRP, soft electrodes are engineered and they are implanted at L3 and L4 dorsal root ganglions (DRGs). ES delivered at DRGs for the first two weeks after fracture increases CGRP expression in both DRGs and fracture callus. It is also identified that CGRP is indispensable for type-H vessel formation, a biological event coupling angiogenesis and osteogenesis, contributing to ES-enhanced osteoporotic fracture healing. This proof-of-concept study shows for the first time that ES at lumbar DRGs can effectively promote femoral fracture healing, offering an innovative strategy using bioelectronic device to enhance bone regeneration.
Collapse
Affiliation(s)
- Jie Mi
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of OrthopaedicsShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine639 Zhizaoju RoadShanghai200011People's Republic of China
| | - Jian‐Kun Xu
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Zhi Yao
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Hao Yao
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Ye Li
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Xuan He
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Bing‐Yang Dai
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Li Zou
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Wen‐Xue Tong
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Xiao‐Tian Zhang
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Pei‐Jie Hu
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Ye Chun Ruan
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Ning Tang
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Xia Guo
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of OrthopaedicsShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine639 Zhizaoju RoadShanghai200011People's Republic of China
| | - Ju‐Fang He
- Departments of Neuroscience and Biomedical SciencesCity University of Hong KongKowloon Tong999077Hong Kong
| | - Ling Qin
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| |
Collapse
|
26
|
Wang C, Zhang Q, Wang R, Xu L. Botulinum Toxin Type A for Diabetic Peripheral Neuropathy Pain: A Systematic Review and Meta-Analysis. J Pain Res 2021; 14:3855-3863. [PMID: 34938114 PMCID: PMC8687679 DOI: 10.2147/jpr.s340390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/07/2021] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Botulinum toxin type A (BTX-A) has been proposed as a treatment for painful diabetic peripheral neuropathy (DPN). This systematic review and meta-analysis aimed to assess the effect and safety of BTX-A for treating DPN pain. METHODS PubMed, Embase, and Cochrane Library were searched for relevant articles published up to July 7, 2021. Randomized clinical trials (RCTs) were included if they were related to the treatment of DPN pain with BTX-A. The primary outcome was the change in intensity of pain and secondary outcomes were adverse effects and changes in sleep and life quality. RESULTS A total of four studies, comprising 231 patients, were included in our systematic review. BTX-A treatment induced a greater reduction in the visual analog scale score (mean difference = -2.52, 95% confidence interval [CI] [-3.06, -1.99], p < 0.001) than did the placebo treatment, with no significant heterogeneity between studies (I2 = 0). BTX-A treatment improved several neuropathy pain scale items (eg, hot sensation, sensitive sensation, unpleasant sensation, deep pain, and surface pain) significantly more than with placebo treatment (p < 0.05 for all). There was no significant difference in adverse effect (relative risk = 1.00, 95% CI [0.97, 1.03], p = 0.89). CONCLUSION Intradermal BTX-A injection was shown to be effective and safe in relieving DPN pain. Further larger scale and well-designed RCTs are needed.
Collapse
Affiliation(s)
- Chengbing Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Qian Zhang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Renjie Wang
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Lei Xu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
27
|
Kim YJ, Granstein RD. Roles of calcitonin gene-related peptide in the skin, and other physiological and pathophysiological functions. Brain Behav Immun Health 2021; 18:100361. [PMID: 34746878 PMCID: PMC8551410 DOI: 10.1016/j.bbih.2021.100361] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 01/05/2023] Open
Abstract
Skin immunity is regulated by many mediator molecules. One is the neuropeptide calcitonin gene-related peptide (CGRP). CGRP has roles in regulating the function of components of the immune system including T cells, B cells, dendritic cells (DCs), endothelial cells (ECs), and mast cells (MCs). Herein we discuss actions of CGRP in mediating inflammatory and vascular effects in various cutaneous models and disorders. CGRP can help to recruit immune cells through endothelium-dependent vasodilation. CGRP plays an important role in the pathogenesis of neurogenic inflammation. Functions of many components in the immune system are influenced by CGRP. CGRP regulates various inflammatory processes in human skin by affecting different cell-types.
Collapse
Affiliation(s)
- Yee Jung Kim
- Department of Dermatology, Weill Cornell Medicine, 1305 York Avenue, WGC9, New York, NY, 10021, USA
| | - Richard D Granstein
- Department of Dermatology, Weill Cornell Medicine, 1305 York Avenue, WGC9, New York, NY, 10021, USA
| |
Collapse
|
28
|
de-la-Hoz JL, de-Pedro M, Martín-Fontelles I, Mesa-Jimenez J, Chivato T, Bagües A. Efficacy of botulinum toxin type A in the management of masticatory myofascial pain: A retrospective clinical study. J Am Dent Assoc 2021; 153:683-691. [PMID: 34763815 DOI: 10.1016/j.adaj.2021.07.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 07/15/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Muscular pain is the main cause of disability worldwide. Myofascial pain of orofacial origin is a frequent condition, the treatment of which is not always accomplished with traditional treatment. Botulinum toxin type A (BTA) is being studied for the treatment of this type of pain with contradicting results. Thus, the objective of this study was to assess the efficacy of BTA in the therapeutic management of masticatory myofascial pain (MFP). CASE DESCRIPTION A retrospective study of 100 patients with a diagnosis of MFP was conducted. The control group (50 patients) received conventional treatment (prescription of a muscle relaxant and craniocervical physical therapy). The BTA group (50 patients) received this same treatment and the infiltration of 100 units of BTA in the masticatory musculature. Subjective and objective pain ratings and range of mandibular movements were recorded before and after the treatment. No differences were found between groups in the baseline values. Statistically significant improvements were found in both groups compared with baseline values in all studied parameters. Moreover, BTA improved the subjective pain ratings compared with the control group. The administration of BTA added to the conventional treatment does not seem to improve objective pain ratings and functional measurements, but it improves the subjective pain ratings. PRACTICAL IMPLICATIONS The addition of BTA could be beneficial in the treatment of MFP in addition to conventional treatment, but further studies are needed to elucidate the mechanisms underlying this positive effect.
Collapse
|
29
|
Sitnikova V, Kämppi A, Teronen O, Kemppainen P. Comprehensive evaluation of botulinum toxin treatment outcomes of a patient with persistent myofascial orofacial pain. Clin Case Rep 2021; 9:e04731. [PMID: 34484765 PMCID: PMC8405429 DOI: 10.1002/ccr3.4731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/28/2021] [Accepted: 08/09/2021] [Indexed: 01/09/2023] Open
Abstract
The outcome evaluation method presented in this case study, including Axes I and II findings combined with the results of quantitative bite force and EMG measurements, provides a good tool for proper evaluation of the effect of BoNT-A on patients with myofascial orofacial pain and changes in jaw muscle function.
Collapse
Affiliation(s)
- Victoria Sitnikova
- Department of Oral and Maxillofacial DiseasesFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Antti Kämppi
- Department of Oral and Maxillofacial DiseasesFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Olli Teronen
- Department of Oral and Maxillofacial DiseasesFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Private Practice MehiläinenHelsinkiFinland
| | - Pentti Kemppainen
- Department of Oral and Maxillofacial DiseasesFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Helsinki University Central Hospital (HUCH)HelsinkiFinland
| |
Collapse
|
30
|
Selective Expression of a SNARE-Cleaving Protease in Peripheral Sensory Neurons Attenuates Pain-Related Gene Transcription and Neuropeptide Release. Int J Mol Sci 2021; 22:ijms22168826. [PMID: 34445536 PMCID: PMC8396265 DOI: 10.3390/ijms22168826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/08/2021] [Accepted: 08/12/2021] [Indexed: 12/31/2022] Open
Abstract
Chronic pain is a leading health and socioeconomic problem and an unmet need exists for long-lasting analgesics. SNAREs (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) are required for neuropeptide release and noxious signal transducer surface trafficking, thus, selective expression of the SNARE-cleaving light-chain protease of botulinum neurotoxin A (LCA) in peripheral sensory neurons could alleviate chronic pain. However, a safety concern to this approach is the lack of a sensory neuronal promoter to prevent the expression of LCA in the central nervous system. Towards this, we exploit the unique characteristics of Pirt (phosphoinositide-interacting regulator of TRP), which is expressed in peripheral nociceptive neurons. For the first time, we identified a Pirt promoter element and cloned it into a lentiviral vector driving transgene expression selectively in peripheral sensory neurons. Pirt promoter driven-LCA expression yielded rapid and concentration-dependent cleavage of SNAP-25 in cultured sensory neurons. Moreover, the transcripts of pain-related genes (TAC1, tachykinin precursor 1; CALCB, calcitonin gene-related peptide 2; HTR3A, 5-hydroxytryptamine receptor 3A; NPY2R, neuropeptide Y receptor Y2; GPR52, G protein-coupled receptor 52; SCN9A, sodium voltage-gated channel alpha subunit 9; TRPV1 and TRPA1, transient receptor potential cation channel subfamily V member 1 and subfamily A member 1) in pro-inflammatory cytokines stimulated sensory neurons were downregulated by viral mediated expression of LCA. Furthermore, viral expression of LCA yielded long-lasting inhibition of pain mediator release. Thus, we show that the engineered Pirt-LCA virus may provide a novel means for long lasting pain relief.
Collapse
|
31
|
Giovannetti O, Tomalty D, Gaudet D, Clohosey D, Forster A, Monaghan M, Harvey MA, Johnston S, Komisaruk B, Goldstein S, Hannan J, Goldstein I, Adams MA. Immunohistochemical Investigation of Autonomic and Sensory Innervation of Anterior Vaginal Wall Female Periurethral Tissue: A Study of the Surgical Field of Mid-Urethral Sling Surgery Using Cadaveric Simulation. J Sex Med 2021; 18:1167-1180. [PMID: 37057425 DOI: 10.1016/j.jsxm.2021.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/26/2021] [Accepted: 05/03/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Female sexual dysfunction, including female orgasm disorder, has been reported following mid-urethral sling (MUS) surgery to treat bothersome stress urinary incontinence. Anterior vaginal wall-female periurethral tissue (AVW-FPT) likely contains autonomic and sensory innervation involved in the female sexual response, and injury to these nerves may result from MUS implantation. AIM To characterize, using fresh cadaveric tissue, autonomic and sensory nerves in AVW- FPT using immunohistochemistry (IHC), and to assess their proximity to an implanted MUS. METHODS AVW-FPT was excised following careful dissection from four fresh cadavers. Prior to dissection, one cadaver underwent simulation of the MUS procedure by a urogynegologist, using a fascial sling. All samples were paraffin embedded, sectioned, and stained with hematoxylin. Serial sectioning and IHC were performed to identify nerves. IHC markers were used to characterize the sensory and autonomic innervation. OUTCOMES IHC localization of autonomic and sensory nerve markers consistent with neural tissue within the region of MUS implantation. RESULTS IHC of AVW-FPT using protein gene product 9.5 (PGP9.5), a general nerve stain, revealed innervation throughout the region targeted by the MUS implantation. More specifically, immunoreactivity for both autonomic (tyrosine hydroxylase, TH) and sensory (Nav1.8 and S100ß) nerves were found in close proximity (<1 mm) to the implanted MUS. In addition, a subset of S100ß positive nerves also showed immunoreactivity for calcitonin gene-related peptide (CGRP). Combining the IHC findings with the surgical simulation of the MUS implantation revealed the potential for damage to both autonomic and sensory nerves as a direct result of the MUS procedure. CLINICAL TRANSLATION The identified autonomic and sensory nerves of the AVW-FPT may contribute to the female sexual response, and yet are potentially negatively impacted by MUS procedures. Given that surgeries performed on male genital tissue, including the prostate, may cause sexual dysfunction secondary to nerve damage, and that urologists routinely provide informed consent regarding this possibility, urogynaecologists are encouraged to obtain appropriate informed consent from prospective patients undergoing the MUS procedure. STRENGTHS & LIMITATIONS This is the first study to characterize the sensory and autonomic innervation within the surgical field of MUS implantation and demonstrate its relationship to an implanted MUS. The small sample size is a limitation of this study. CONCLUSION The present study provides evidence of potential injury to autonomic and sensory innervation of AVW-FPT as a consequence of MUS implantation, which may help explain the underlying mechanisms involved in the reported post-operative female sexual dysfunction in some women. Giovannetti O, Tomalty D, Gaudet D, et al. Immunohistochemical Investigation of Autonomic and Sensory Innervation of Anterior Vaginal Wall Female Periurethral Tissue: A Study of the Surgical Field of Mid-Urethral Sling Surgery Using Cadaveric Simulation. J Sex Med 2021;18:1168-1180.
Collapse
|
32
|
Botulinum toxin A in Patients with Chronic Migraine: A Single-Center Experience;. JOURNAL OF CONTEMPORARY MEDICINE 2021. [DOI: 10.16899/jcm.821703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
33
|
de Paola M, Garrido F, Zanetti MN, Michaut MA. VAMPs sensitive to tetanus toxin are required for cortical granule exocytosis in mouse oocytes. Exp Cell Res 2021; 405:112629. [PMID: 34023392 DOI: 10.1016/j.yexcr.2021.112629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 01/16/2023]
Abstract
Fusion of cortical granules with oocyte plasma membrane is one of the most significant secretory events to prevent polyspermy during oocyte activation. Cortical granule exocytosis (CGE) is distinct from most other exocytosis because cortical granules are not renewed after secretion. However, it is thought to be mediated by SNARE complex, which mediates membrane fusion in other exocytoses. SNAREs proteins are divided into Q (glutamine)- and R (arginine)-SNAREs. Q-SNAREs include Syntaxins and SNAP25 family, and R-SNAREs include VAMPs family. In mouse oocytes, Syntaxin4 and SNAP23 have been involved in CGE; nevertheless, it is unknown if VAMP is required. Here, we demonstrated by RT-PCR and immunoblotting that VAMP1 and VAMP3 are expressed in mouse oocyte, and they localized in the cortical region of this cell. Using a functional assay to quantify CGE, we showed that tetanus toxin -which specifically cleavages VAMP1, VAMP2 or VAMP3- inhibited CGE suggesting that at least one VAMP was necessary. Function blocking assays demonstrated that only the microinjection of anti-VAMP1 or anti-VAMP3 antibodies abolished CGE in activated oocytes. These findings demonstrate that R-SNAREs sensitive to tetanus toxin, VAMP1 and VAMP3 -but not VAMP2-, are required for CGE and demonstrate that CGE is mediated by the SNARE complex.
Collapse
Affiliation(s)
- Matilde de Paola
- Laboratorio de Biología Reproductiva y Molecular, Instituto de Histología y Embriología, Universidad Nacional de Cuyo-CONICET, Av. Libertador 80, 5500, Mendoza, Argentina; Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Av. Libertador 80, 5500, Mendoza, Argentina
| | - Facundo Garrido
- Laboratorio de Biología Reproductiva y Molecular, Instituto de Histología y Embriología, Universidad Nacional de Cuyo-CONICET, Av. Libertador 80, 5500, Mendoza, Argentina
| | - María N Zanetti
- Laboratorio de Biología Reproductiva y Molecular, Instituto de Histología y Embriología, Universidad Nacional de Cuyo-CONICET, Av. Libertador 80, 5500, Mendoza, Argentina
| | - Marcela Alejandra Michaut
- Laboratorio de Biología Reproductiva y Molecular, Instituto de Histología y Embriología, Universidad Nacional de Cuyo-CONICET, Av. Libertador 80, 5500, Mendoza, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Padre Jorge Contreras, 1300, Mendoza, Argentina.
| |
Collapse
|
34
|
Affiliation(s)
- Haya S. Raef
- Tufts University School of Medicine, Boston, Massachusetts
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sarina B. Elmariah
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Topical Treatments and Their Molecular/Cellular Mechanisms in Patients with Peripheral Neuropathic Pain-Narrative Review. Pharmaceutics 2021; 13:pharmaceutics13040450. [PMID: 33810493 PMCID: PMC8067282 DOI: 10.3390/pharmaceutics13040450] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 12/25/2022] Open
Abstract
Neuropathic pain in humans results from an injury or disease of the somatosensory nervous system at the peripheral or central level. Despite the considerable progress in pain management methods made to date, peripheral neuropathic pain significantly impacts patients' quality of life, as pharmacological and non-pharmacological methods often fail or induce side effects. Topical treatments are gaining popularity in the management of peripheral neuropathic pain, due to excellent safety profiles and preferences. Moreover, topical treatments applied locally may target the underlying mechanisms of peripheral sensitization and pain. Recent studies showed that peripheral sensitization results from interactions between neuronal and non-neuronal cells, with numerous signaling molecules and molecular/cellular targets involved. This narrative review discusses the molecular/cellular mechanisms of drugs available in topical formulations utilized in clinical practice and their effectiveness in clinical studies in patients with peripheral neuropathic pain. We searched PubMed for papers published from 1 January 1995 to 30 November 2020. The key search phrases for identifying potentially relevant articles were "topical AND pain", "topical AND neuropathic", "topical AND treatment", "topical AND mechanism", "peripheral neuropathic", and "mechanism". The result of our search was 23 randomized controlled trials (RCT), 9 open-label studies, 16 retrospective studies, 20 case (series) reports, 8 systematic reviews, 66 narrative reviews, and 140 experimental studies. The data from preclinical studies revealed that active compounds of topical treatments exert multiple mechanisms of action, directly or indirectly modulating ion channels, receptors, proteins, and enzymes expressed by neuronal and non-neuronal cells, and thus contributing to antinociception. However, which mechanisms and the extent to which the mechanisms contribute to pain relief observed in humans remain unclear. The evidence from RCTs and reviews supports 5% lidocaine patches, 8% capsaicin patches, and botulinum toxin A injections as effective treatments in patients with peripheral neuropathic pain. In turn, single RCTs support evidence of doxepin, funapide, diclofenac, baclofen, clonidine, loperamide, and cannabidiol in neuropathic pain states. Topical administration of phenytoin, ambroxol, and prazosin is supported by observational clinical studies. For topical amitriptyline, menthol, and gabapentin, evidence comes from case reports and case series. For topical ketamine and baclofen, data supporting their effectiveness are provided by both single RCTs and case series. The discussed data from clinical studies and observations support the usefulness of topical treatments in neuropathic pain management. This review may help clinicians in making decisions regarding whether and which topical treatment may be a beneficial option, particularly in frail patients not tolerating systemic pharmacotherapy.
Collapse
|
36
|
Abstract
Background and objectives Botulinum toxin injection has been used for many years for various indications such as cervical dystonia, blepharospasm, oromandibular dystonia, hemifacial spasm in neurology. Botulinum toxin injections have been made in our clinic for about ten years. We want to report our experiences about botulinum toxin treatment in neurologic disease. Methods In this study, the data of the patients receiving BoNT injection between January 2018 and December 2019 was retrospectively analyzed. Age, gender, botulinum toxin indications, and mean dose, duration of efficacy, side effects noted for each injection were recorded. Results There were 122 patients who received botulinum toxin injections between January 2018 and December 2019. Of the 122 patients identified, 28 had cervical dystonia, 61 had HFS, 21 had blepharospasm, 4 had generalized dystonia, 1 had hemidystonia following thalamic bleeding, 1 had tardive dystonia, 4 had migraine, 1 had bruxism and 1 had both migraine and bruxism. Conclusion BoNT injection is a treatment that has been used for various indications in neurology for almost 40 years. Side effects are limited and temporary with appropriate injections. We also had a wide range of indication profiles and high numbers of patients to whom we administered the BoNT treatment.
Collapse
|
37
|
Chang KV, Chiu YH, Wu WT, Hsu PC, Özçakar L. Botulinum toxin injections for shoulder and upper limb pain: a narrative review. Pain Manag 2020; 10:411-420. [PMID: 33073703 DOI: 10.2217/pmt-2020-0015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Botulinum toxin (BoNT) has been widely employed to treat poststroke spasticity, cervical dystonia and muscle hyperactivity. Recently, BoNT injections are increasingly used in treating musculoskeletal pain. The mechanism of BoNT in pain relief comprises relaxation of overused muscles and inhibition of inflammatory nociceptive cytokines/neurotransmitters. As BoNT injections seem promising in treating painful musculoskeletal disorders, we aimed to investigate its effectiveness in shoulder and upper limb pain. Although the present article is a narrative review, we employed a systematic approach to search for relevant articles in PubMed. A total of 19 clinical studies were included. Here, we observed that intramuscular BoNT injections were helpful in stroke patients with hemiplegic shoulder pain. In shoulder joint pain, intra-articular and intrabursal BoNT injections achieved a longer period of pain relief than corticosteroid injections. Similarly, a more durable effect of intramuscular BoNT than saline injections was seen in shoulder myofascial pain. Its use in complex regional pain syndrome and persistent upper limb pain in breast cancer survivors was insufficient, necessitating more studies. Since not all of the included studies could provide Class I of evidence based on the efficacy criteria used by American Academy of Neurology, controlled clinical trials in a larger number of patients are necessary to verify validity of these findings in the future.
Collapse
Affiliation(s)
- Ke-Vin Chang
- Department of Physical Medicine & Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei, 10845, Taiwan.,Department of Physical Medicine & Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, 10048, Taiwan
| | - Yi-Hsiang Chiu
- Department of Physical Medicine & Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei, 10845, Taiwan
| | - Wei-Ting Wu
- Department of Physical Medicine & Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei, 10845, Taiwan.,Department of Physical Medicine & Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, 10048, Taiwan
| | - Po-Cheng Hsu
- Department of Physical Medicine & Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, 10048, Taiwan
| | - Levent Özçakar
- Department of Physical & Rehabilitation Medicine, Hacettepe University Medical School, Ankara, 06100, Turkey
| |
Collapse
|
38
|
Chen ST, Wu JW. A new era for migraine: The role of calcitonin gene-related peptide in the trigeminovascular system. PROGRESS IN BRAIN RESEARCH 2020; 255:123-142. [PMID: 33008504 DOI: 10.1016/bs.pbr.2020.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/26/2020] [Accepted: 05/01/2020] [Indexed: 03/03/2023]
Abstract
There is a huge improvement in our understanding of migraine pathophysiology in the past decades. The activation of the trigeminovascular system has been proved to play a key role in migraine. Calcitonin gene-related peptide (CGRP) and CGRP receptors are widely distributed in the trigeminovascular system. The CGRP is expressed on the C-fibers, and the CGRP receptors are distributed on the A-δ fibers of the trigeminal ganglion and nerves. Further studies found elevated serum CGRP level during migraine attacks, and infusion of CGRP can trigger migraine-like attacks, provide more direct evidence of the link between CGRP and migraine attack. Based on these findings, several treatment options have been designed for migraine treatment, including CGRP receptor antagonists (gepants) and monoclonal antibodies targeting CGRP or CGRP receptors. The clinical trials show both gepants and monoclonal antibodies are effective for migraine treatment. In this section, we describe the roles of the trigeminovascular system in migraine, the discovery of CGRP, and the CGRP signaling pathway.
Collapse
Affiliation(s)
- Shu-Ting Chen
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jr-Wei Wu
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan; Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.
| |
Collapse
|
39
|
Drinovac Vlah V, Bach-Rojecky L. What have we learned about antinociceptive effect of botulinum toxin type A from mirror-image pain models? Toxicon 2020; 185:164-173. [DOI: 10.1016/j.toxicon.2020.07.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/06/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022]
|
40
|
Hoogstraaten RI, van Keimpema L, Toonen RF, Verhage M. Tetanus insensitive VAMP2 differentially restores synaptic and dense core vesicle fusion in tetanus neurotoxin treated neurons. Sci Rep 2020; 10:10913. [PMID: 32616842 PMCID: PMC7331729 DOI: 10.1038/s41598-020-67988-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 06/11/2020] [Indexed: 01/10/2023] Open
Abstract
The SNARE proteins involved in the secretion of neuromodulators from dense core vesicles (DCVs) in mammalian neurons are still poorly characterized. Here we use tetanus neurotoxin (TeNT) light chain, which cleaves VAMP1, 2 and 3, to study DCV fusion in hippocampal neurons and compare the effects on DCV fusion to those on synaptic vesicle (SV) fusion. Both DCV and SV fusion were abolished upon TeNT expression. Expression of tetanus insensitive (TI)-VAMP2 restored SV fusion in the presence of TeNT, but not DCV fusion. Expression of TI-VAMP1 or TI-VAMP3 also failed to restore DCV fusion. Co-transport assays revealed that both TI-VAMP1 and TI-VAMP2 are targeted to DCVs and travel together with DCVs in neurons. Furthermore, expression of the TeNT-cleaved VAMP2 fragment or a protease defective TeNT in wild type neurons did not affect DCV fusion and therefore cannot explain the lack of rescue of DCV fusion by TI-VAMP2. Finally, to test if two different VAMPs might both be required in the DCV secretory pathway, Vamp1 null mutants were tested. However, VAMP1 deficiency did not reduce DCV fusion. In conclusion, TeNT treatment combined with TI-VAMP2 expression differentially affects the two main regulated secretory pathways: while SV fusion is normal, DCV fusion is absent.
Collapse
Affiliation(s)
- Rein I Hoogstraaten
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit (VU) Amsterdam and University Medical Center Amsterdam, de Boelelaan 1087, 1018 HV, Amsterdam, The Netherlands
| | - Linda van Keimpema
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit (VU) Amsterdam and University Medical Center Amsterdam, de Boelelaan 1087, 1018 HV, Amsterdam, The Netherlands
- Sylics (Synaptologics BV), PO Box 71033, 1008 BA, Amsterdam, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit (VU) Amsterdam and University Medical Center Amsterdam, de Boelelaan 1087, 1018 HV, Amsterdam, The Netherlands.
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit (VU) Amsterdam and University Medical Center Amsterdam, de Boelelaan 1087, 1018 HV, Amsterdam, The Netherlands.
- Clinical Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit (VU) Amsterdam and University Medical Center Amsterdam, de Boelelaan 1087, 1018 HV, Amsterdam, The Netherlands.
| |
Collapse
|
41
|
Mecklenburg J, Sanchez Del Rio M, Reuter U. Cluster headache therapies: pharmacology and mode of action. Expert Rev Clin Pharmacol 2020; 13:641-654. [DOI: 10.1080/17512433.2020.1774361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Jasper Mecklenburg
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Uwe Reuter
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
42
|
Pellesi L, Do TP, Ashina H, Ashina M, Burstein R. Dual Therapy With Anti-CGRP Monoclonal Antibodies and Botulinum Toxin for Migraine Prevention: Is There a Rationale? Headache 2020; 60:1056-1065. [PMID: 32437038 DOI: 10.1111/head.13843] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/29/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To narratively review the pathophysiological rationale of dual therapy with anti-calcitonin gene-related peptide monoclonal antibodies and botulinum toxin type A in treatment-resistant chronic migraine prevention. BACKGROUND For the prevention of chronic migraine, several pharmacological therapies are available, including oral medications, botulinum toxin type A, and the newly approved monoclonal antibodies targeting calcitonin gene-related peptide or its receptor. However, monotherapy does not yield benefits in some affected individuals, which raises the question of whether dual therapy with monoclonal antibodies and botulinum toxin type A hold promise in patients with treatment-resistant chronic migraine. METHOD We searched MEDLINE for articles published from database inception to December 31st, 2019. Publications were largely selected from the past 10 years but commonly referenced and highly regarded older publications were not excluded. RESULTS Preclinical data suggest that anti-calcitonin gene-related peptide monoclonal antibodies and botulinum toxin type A have synergistic effects within the trigeminovascular system. Of note, findings indicate that fremanezumab - an antibody targeting the calcitonin gene-related peptide - mainly prevents the activation of Aδ-fibers, whereas botulinum toxin type A prevents the activation of C-fibers. CONCLUSION There is currently only indirect preclinical evidence to support a rationale for dual therapy with anti-calcitonin gene-related peptide monoclonal antibodies and botulinum toxin type A for chronic migraine prevention. Rigorous studies evaluating clinical efficacy, safety, and cost-effectiveness are needed.
Collapse
Affiliation(s)
- Lanfranco Pellesi
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thien P Do
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Håkan Ashina
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Messoud Ashina
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rami Burstein
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
Blanshan N, Krug H. The Use of Botulinum Toxin for the Treatment of Chronic Joint Pain: Clinical and Experimental Evidence. Toxins (Basel) 2020; 12:toxins12050314. [PMID: 32397671 PMCID: PMC7291335 DOI: 10.3390/toxins12050314] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/25/2020] [Accepted: 05/05/2020] [Indexed: 12/11/2022] Open
Abstract
Chronic osteoarthritis pain is an increasing worldwide problem. Treatment for osteoarthritis pain is generally inadequate or fraught with potential toxicities. Botulinum toxins (BoNTs) are potent inhibitors of neuropeptide release. Paralytic toxicity is due to inhibition at the neuromuscular junction, and this effect has been utilized for treatments of painful dystonias. Pain relief following BoNT muscle injection has been noted to be more significant than muscle weakness and hypothesized to occur because of the inhibition of peripheral neuropeptide release and reduction of peripheral sensitization. Because of this observation, BoNT has been studied as an intra-articular (IA) analgesic for chronic joint pain. In clinical trials, BoNT appears to be effective for nociceptive joint pain. No toxicity has been reported. In preclinical models of joint pain, BoNT is similarly effective. Examination of the dorsal root ganglion (DRG) and the central nervous system has shown that catalytically active BoNT is retrogradely transported by neurons and then transcytosed to afferent synapses in the brain. This suggests that pain relief may also be due to the central effects of the drug. In summary, BoNT appears to be safe and effective for the treatment of chronic joint pain. The long-term effects of IA BoNT are still being determined.
Collapse
Affiliation(s)
- Nicole Blanshan
- Minneapolis VA Health Care System, Minneapolis, MN 55455, USA;
| | - Hollis Krug
- Minneapolis VA Health Care System, Minneapolis, MN 55455, USA;
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
- Correspondence: ; Tel.: +1-612-467-4190
| |
Collapse
|
44
|
The Use of Botulinum Toxin in Pain Management: Basic Science and Clinical Applications. Plast Reconstr Surg 2020; 145:629e-636e. [DOI: 10.1097/prs.0000000000006559] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
45
|
Cost-Effectiveness of Long-Term, Targeted OnabotulinumtoxinA versus Peripheral Trigger Site Deactivation Surgery for the Treatment of Refractory Migraine Headaches. Plast Reconstr Surg 2020; 145:401e-406e. [DOI: 10.1097/prs.0000000000006480] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Tang M, Meng J, Wang J. New Engineered-Botulinum Toxins Inhibit the Release of Pain-Related Mediators. Int J Mol Sci 2019; 21:ijms21010262. [PMID: 31906003 PMCID: PMC6981458 DOI: 10.3390/ijms21010262] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/20/2019] [Accepted: 12/27/2019] [Indexed: 12/11/2022] Open
Abstract
Targeted delivery of potent inhibitor of cytokine/pain-mediator into inflammatory or pain-sensing cells is a promising avenue for treating chronic pain, a world-wide major healthcare burden. An unmet need exists for a specific and effective delivery strategy. Herein, we describe a new approach using sortase to site-specifically ligate a non-toxic botulinum neurotoxin D (BoNT/D) core-therapeutic (synaptobrevin-cleaving protease and translocation domains) to cell-specific targeting ligands. An engineered core-therapeutic was efficiently ligated to IL-1β ligand within minutes. The resultant conjugate specifically entered into cultured murine primary macrophages, cleaved synaptobrevin 3 and inhibited LPS/IFN-γ evoked IL-6 release. Likewise, a CGRP receptor antagonist ligand delivered BoNT/D protease into sensory neurons and inhibited K+-evoked substance P release. As cytokines and neuropeptides are major regulators of inflammation and pain, blocking their release by novel engineered inhibitors highlights their therapeutic potential. Our report describes a new and widely-applicable strategy for the production of targeted bio-therapeutics for numerous chronic diseases.
Collapse
Affiliation(s)
| | - Jianghui Meng
- Correspondence: (J.M.); (J.W.); Tel.: +353-1700-7351 (J.M.); +353-1700-7489 (J.W.)
| | - Jiafu Wang
- Correspondence: (J.M.); (J.W.); Tel.: +353-1700-7351 (J.M.); +353-1700-7489 (J.W.)
| |
Collapse
|
47
|
Khan FA, Mohammed AE, Poongkunran M, Chimakurthy A, Pepper M. Wearing Off Effect of OnabotulinumtoxinA Near the End of Treatment Cycle for Chronic Migraine: A 4-Year Clinical Experience. Headache 2019; 60:430-440. [PMID: 31758548 DOI: 10.1111/head.13713] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2019] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The injection interval for onabotulinumtoxinA (BoNTA) in the management of chronic migraine (CM) is 12 weeks (78-84 days). The aim of this study was to review patient-reported wearing off effect (WOE) of the therapeutic benefit of BoNTA near the end of the treatment cycle. We intended to describe the demographics of patients at baseline and compare groups of patients with multiple episodes of WOE. METHODS We conducted a retrospective review of patients with CM who received uninterrupted BoNTA therapy from January 2014 to March 2018. The data from patient-reported WOE (worsening headache variables and neck pain) that occurred during the 4 weeks (28 days) prior to the scheduled re-injection of BoNTA for treatment cycles with injection interval ≤13 weeks and without obvious confounding factors were reviewed. RESULTS We identified 98 eligible patients and analyzed 471 treatment cycles. Forty-three unique patients reported at least 1 occurrence of WOE. About 24/43 patients reported 1 WOE event and 19/43 patients reported ≥2 WOE events. Between the 2 groups, anxiety disorder and opioid use for headache were statistically significantly different. In the former group, the median interquartile range (IQR) dose of BoNTA was 165 (155, 175) units and the median IQR duration of the antinociceptive effect of BoNTA was 66.5 (63, 71.5) days. In the latter group, the median IQR dose of BoNTA was 167 (155, 173.3) units and the median IQR duration of the antinociceptive effect of BoNTA was 65.3 (62.5, 68.8) days. Up to 32% of these patients reported an increase in the use of abortive therapies to manage the symptoms of WOE. DISCUSSION The primary goal of BoNTA in the treatment of CM is to mitigate the development of central sensitization. Since the 12-week injection paradigm may not provide sustained antinociceptive effect in all patients, it may account for the failure of response to BoNTA. Repeated occurrences of the WOE can potentially lead to medication overuse and impact quality of life.
Collapse
Affiliation(s)
- Fawad A Khan
- McCasland Family Comprehensive Headache Center, Ochsner Neuroscience Institute, Ochsner Clinic Foundation, New Orleans, LA, USA.,The University of Queensland School of Medicine, Ochsner Clinical School, New Orleans, LA, USA.,Tulane University School of Medicine, New Orleans, LA, USA
| | - Alaa E Mohammed
- Center for Outcomes and Health Services Research, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - Mugilan Poongkunran
- McCasland Family Comprehensive Headache Center, Ochsner Neuroscience Institute, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - Anilkumar Chimakurthy
- McCasland Family Comprehensive Headache Center, Ochsner Neuroscience Institute, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - Michael Pepper
- McCasland Family Comprehensive Headache Center, Ochsner Neuroscience Institute, Ochsner Clinic Foundation, New Orleans, LA, USA.,The University of Queensland School of Medicine, Ochsner Clinical School, New Orleans, LA, USA
| |
Collapse
|
48
|
Matak I, Bölcskei K, Bach-Rojecky L, Helyes Z. Mechanisms of Botulinum Toxin Type A Action on Pain. Toxins (Basel) 2019; 11:E459. [PMID: 31387301 PMCID: PMC6723487 DOI: 10.3390/toxins11080459] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/31/2022] Open
Abstract
Already a well-established treatment for different autonomic and movement disorders, the use of botulinum toxin type A (BoNT/A) in pain conditions is now continuously expanding. Currently, the only approved use of BoNT/A in relation to pain is the treatment of chronic migraines. However, controlled clinical studies show promising results in neuropathic and other chronic pain disorders. In comparison with other conventional and non-conventional analgesic drugs, the greatest advantages of BoNT/A use are its sustained effect after a single application and its safety. Its efficacy in certain therapy-resistant pain conditions is of special importance. Novel results in recent years has led to a better understanding of its actions, although further experimental and clinical research is warranted. Here, we summarize the effects contributing to these advantageous properties of BoNT/A in pain therapy, specific actions along the nociceptive pathway, consequences of its central activities, the molecular mechanisms of actions in neurons, and general pharmacokinetic parameters.
Collapse
Affiliation(s)
- Ivica Matak
- Department of Pharmacology, University of Zagreb School of Medicine, Šalata 11, 10000 Zagreb, Croatia.
| | - Kata Bölcskei
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, 7624 Pécs, Hungary
- János Szentágothai Research Center, Center for Neuroscience, University of Pécs, Ifjúság útja 20, 7624 Pécs, Hungary
| | - Lidija Bach-Rojecky
- Department of Pharmacology, University of Zagreb Faculty of Pharmacy and Biochemistry, Domagojeva 2, 10000 Zagreb, Croatia
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, 7624 Pécs, Hungary
- János Szentágothai Research Center, Center for Neuroscience, University of Pécs, Ifjúság útja 20, 7624 Pécs, Hungary
| |
Collapse
|
49
|
Kumar A, Potts JD, DiPette DJ. Protective Role of α-Calcitonin Gene-Related Peptide in Cardiovascular Diseases. Front Physiol 2019; 10:821. [PMID: 31312143 PMCID: PMC6614340 DOI: 10.3389/fphys.2019.00821] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/11/2019] [Indexed: 01/09/2023] Open
Abstract
α-Calcitonin gene-related peptide (α-CGRP) is a regulatory neuropeptide of 37 amino acids. It is widely distributed in the central and peripheral nervous system, predominantly in cell bodies of the dorsal root ganglion (DRG). It is the most potent vasodilator known to date and has inotropic and chronotropic effects. Using pharmacological and genetic approaches, our laboratory and other research groups established the protective role of α-CGRP in various cardiovascular diseases such as heart failure, experimental hypertension, myocardial infarction, and myocardial ischemia/reperfusion injury (I/R injury). α-CGRP acts as a depressor to attenuate the rise in blood pressure in three different models of experimental hypertension: (1) DOC-salt, (2) subtotal nephrectomy-salt, and (3) L-NAME-induced hypertension during pregnancy. Subcutaneous administration of α-CGRP lowers the blood pressure in hypertensive and normotensive humans and rodents. Recent studies also demonstrated that an α-CGRP analog, acylated α-CGRP, with extended half-life (~7 h) reduces blood pressure in Ang-II-induced hypertensive mouse, and protects against abdominal aortic constriction (AAC)-induced heart failure. Together, these studies suggest that α-CGRP, native or a modified form, may be a potential therapeutic agent to treat patients suffering from cardiac diseases.
Collapse
Affiliation(s)
- Ambrish Kumar
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Jay D Potts
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Donald J DiPette
- Department of Internal Medicine, School of Medicine, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
50
|
Abstract
Migraine is a strongly disabling disease characterized by a unilateral throbbing headache lasting for up to 72 h for each individual attack. There have been many theories on the pathophysiology of migraine throughout the years. Currently, the neurovascular theory dominates, suggesting clear involvement of the trigeminovascular system. The most recent data show that a migraine attack most likely originates in the hypothalamus and activates the trigeminal nucleus caudalis (TNC). Although the mechanisms are unknown, activation of the TNC leads to peripheral release of calcitonin gene-related protein (CGRP), most likely from C-fibers. During the past year monoclonal antibodies against CGRP or the CGRP receptor have emerged as the most promising targets for migraine therapy, and at the same time established the strong involvement of CGRP in the pathophysiology of migraine. The viewpoint presented here focuses further on the activation of the CGRP receptor on the sensory Aδ-fiber, leading to the sensation of pain. The CGRP receptor activates adenylate cyclase, which leads to an increase in cyclic adenosine monophosphate (cAMP). We hypothesize that cAMP activates the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, triggering an action potential sensed as pain. The mechanisms behind migraine pain on a molecular level, particularly their importance to cAMP, provide clues to potential new anti-migraine targets. In this article we focus on the development of targets related to the CGRP system, and further include novel targets such as the pituitary adenylate cyclase-activating peptide (PACAP) system, the serotonin 5-HT1F receptor, purinergic receptors, HCN channels, adenosine triphosphate-sensitive potassium channels (KATP), and the glutaminergic system.
Collapse
|