1
|
Stein C. Effects of pH on opioid receptor activation and implications for drug design. Biophys J 2024; 123:4158-4166. [PMID: 38970252 PMCID: PMC11700362 DOI: 10.1016/j.bpj.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024] Open
Abstract
G-protein-coupled receptors are integral membrane proteins that transduce chemical signals from the extracellular matrix into the cell. Traditional drug design has considered ligand-receptor interactions only under normal conditions. However, studies on opioids indicate that such interactions are very different in diseased tissues. In such microenvironments, protons play an important role in structural and functional alterations of both ligands and receptors. The pertinent literature strongly suggests that future drug design should take these aspects into account in order to reduce adverse side effects while preserving desired effects of novel compounds.
Collapse
Affiliation(s)
- Christoph Stein
- Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Experimental Anaesthesiology, Berlin, Germany.
| |
Collapse
|
2
|
Pal B, Ghosh R, Sarkar RD, Roy GS. The irreversible, towards fatalic neuropathy: from the genesis of diabetes. Acta Diabetol 2024:10.1007/s00592-024-02429-4. [PMID: 39636401 DOI: 10.1007/s00592-024-02429-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Diabetic neuropathy is the most prevalent diabetes-associated complication that negatively impacts the quality of life of the patients. The extensive complications of diabetic peoples in the world are the leading cause of neuropathic pain, and over-activation of different biochemical signalling process induces the pathogenic progression and are also corresponding the epidemic painful symptom of diabetic neuropathy. The main prevalent abnormality is neuropathy, which further causing distal symmetric polyneuropathy and focal neuropathy. The exact pathological complication of diabetes associated neuropathic algesia is still unclear, but the alteration in micro-angiopathy associated nerve fibre loss, hyper polyol formation, MAPK signalling, WNT signalling, tau-derived insulin signalling processes are well known. Furthermore, the post-translational modification of different ion channels, oxidative and nitrosative stress, brain plasticity and microvascular changes can contributes the development of neuropathic pain. However, in the current review we discussed about these pathogenic development of neuropathic pain from the genesis of diabetes, and how diabetes affects the physiological and psychological health, and quality of life of the patients. Furthermore, the treatment of diabetic neuropathy with conventional monotherapy and emerging therapy are discussed. In addition, the treatment with phytochemical constituents their mechanisms and clinical evidences are also reported. The future investigation is required on pathological alteration occurs in neuropathic individuals, and on molecular mechanisms as well as the adverse effect of phytochemicals to determine all aspects of neuropathic algesia including effective treatments, which will prevents the sympathetic pain in patients.
Collapse
Affiliation(s)
- Bhaskar Pal
- Department of Pharmacology, Charaktala College of Pharmacy, Charaktala, Mothabari, Malda, West Bengal, India.
| | - Rashmi Ghosh
- Bengal College of Pharmaceutical Science & Research, Durgapur, West Bengal, India
| | - Raktimava Das Sarkar
- Department of Pharmaceutical Technology, Bengal School of Technology, Sugandha, Delhi Road, Chinsurah, Hooghly, West Bengal, India
| | - Gouranga Sundar Roy
- Department of Pharmaceutical Technology, Bengal School of Technology, Sugandha, Delhi Road, Chinsurah, Hooghly, West Bengal, India
| |
Collapse
|
3
|
Rosner J, Attal N, Finnerup NB. Clinical pharmacology of neuropathic pain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:403-430. [PMID: 39580218 DOI: 10.1016/bs.irn.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
This chapter aims to review the current pharmacological options for neuropathic pain treatment, their mechanisms of action, and future directions for clinical practice. Achieving pain relief in neuropathic pain conditions remains a challenge in clinical practice. The field of pharmacotherapy for neuropathic pain has encountered significant difficulties in translating substantial advances in our understanding of the underlying pathophysiological mechanisms into clinically effective therapies. This chapter presents the drugs recommended for the pharmacotherapy of neuropathic pain, based on the widely accepted treatment guidelines formulated by the Neuropathic Pain Special Interest Group of the International Association for the Study of Pain. In addition to discussing how the evidence base is created as part of international consortia, the drugs are also examined in terms of their putative molecular mechanisms as well as pharmacological pleiotropy, i.e., their potential unspecific and multi-target effects resulting in modulation of neuronal hyperexcitability. The chapter closes with a discussion of potential future developments in the field.
Collapse
Affiliation(s)
- Jan Rosner
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland.
| | - Nadine Attal
- Inserm U987, APHP, CHU Ambroise Pare, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
| | - Nanna B Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
4
|
Zurek NA, Thiyagarajan S, Ehsanian R, Goins AE, Goyal S, Shilling M, Lambert CG, Westlund KN, Alles SRA. Machine Learning Elucidates Electrophysiological Properties Predictive of Multi- and Single-Firing Human and Mouse Dorsal Root Ganglia Neurons. eNeuro 2024; 11:ENEURO.0248-24.2024. [PMID: 39299808 PMCID: PMC11457269 DOI: 10.1523/eneuro.0248-24.2024] [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: 06/09/2024] [Revised: 08/08/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024] Open
Abstract
Human and mouse dorsal root ganglia (hDRG and mDRG) neurons are important tools in understanding the molecular and electrophysiological mechanisms that underlie nociception and drive pain behaviors. One of the simplest differences in firing phenotypes is that neurons are single-firing (exhibit only one action potential) or multi-firing (exhibit 2 or more action potentials). To determine if single- and multi-firing hDRG neurons exhibit differences in intrinsic properties, firing phenotypes, and AP waveform properties, and if these properties could be used to predict multi-firing, we measured 22 electrophysiological properties by whole-cell patch-clamp electrophysiology of 94 hDRG neurons from six male and four female donors. We then analyzed the data using several machine learning models to determine if these properties could be used to predict multi-firing. We used 1,000 iterations of Monte Carlo cross-validation to split the data into different train and test sets and tested the logistic regression, k-nearest neighbors, random forest, support vector classifier, and XGBoost machine learning models. All models tested had a >80% accuracy on average, with support vector classifier, and XGBoost performing the best. We found that several properties correlated with multi-firing hDRG neurons and together could be used to predict multi-firing neurons in hDRG including a long decay time, a low rheobase, and long first spike latency. We also found that the hDRG models were able to predict multi-firing with 90% accuracy in mDRG neurons. Understanding these properties could be beneficial in the elucidation of targets on peripheral sensory neurons related to pain.
Collapse
Affiliation(s)
- Nesia A Zurek
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Sherwin Thiyagarajan
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Reza Ehsanian
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Aleyah E Goins
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Sachin Goyal
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Mark Shilling
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Christophe G Lambert
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131
| | - Karin N Westlund
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| | - Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87106
| |
Collapse
|
5
|
Gualdani R, Barbeau S, Yuan JH, Jacobs DS, Gailly P, Dib-Hajj SD, Waxman SG. TRPV1 corneal neuralgia mutation: Enhanced pH response, bradykinin sensitization, and capsaicin desensitization. Proc Natl Acad Sci U S A 2024; 121:e2406186121. [PMID: 39226353 PMCID: PMC11406256 DOI: 10.1073/pnas.2406186121] [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: 03/26/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
The factors that contribute to pain after nerve injury remain incompletely understood. Laser-assisted in situ keratomileusis (LASIK) and photorefractive keratectomy (PRK) are common surgical techniques to correct refractive errors. After LASIK or PRK, a subset of patients suffers intense and persistent pain, of unknown origin, described by patients as feeling like shards of glass in their eye. Here, we evaluated a TRPV1 variant, p.V527M, found in a 49-y-old woman who developed corneal pain after LASIK and subsequent PRK enhancement, reporting an Ocular Surface Disease Index score of 100. Using patch-clamp and Ca2+ imaging, we found that the V527M mutation enhances the response to acidic pH. Increasing proton concentration induced a stronger leftward shift in the activation curve of V527M compared to WT, resulting in channel activity of the mutant in acidic pH at more physiological membrane potentials. Finally, comparing the responses to consecutive applications of different agonists, we found in V527M channels a reduced capsaicin-induced desensitization and increased sensitization by the arachidonic acid metabolite 12-hydroxyeicosatetraenoic acid (12-HETE). We hypothesize that the increased response in V527M channels to protons and enhanced sensitization by 12-HETE, two inflammatory mediators released in the cornea after tissue damage, may contribute to the pathogenesis of corneal neuralgia after refractive surgery.
Collapse
Affiliation(s)
- Roberta Gualdani
- Laboratory of Cell Physiology, Institute of Neuroscience, Université catholique de Louvain, BrusselsB-1200, Belgium
| | - Solène Barbeau
- Laboratory of Cell Physiology, Institute of Neuroscience, Université catholique de Louvain, BrusselsB-1200, Belgium
| | - Jun-Hui Yuan
- Department of Neurology, Yale School of Medicine, New Haven, CT06520
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT06520
- Neurorehabilitation Research Center, Veterans Affairs Medical Center, West Haven, CT06516
| | - Deborah S. Jacobs
- Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA02114
| | - Philippe Gailly
- Laboratory of Cell Physiology, Institute of Neuroscience, Université catholique de Louvain, BrusselsB-1200, Belgium
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale School of Medicine, New Haven, CT06520
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT06520
- Neurorehabilitation Research Center, Veterans Affairs Medical Center, West Haven, CT06516
| | - Stephen G. Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT06520
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, New Haven, CT06520
- Neurorehabilitation Research Center, Veterans Affairs Medical Center, West Haven, CT06516
| |
Collapse
|
6
|
Philibert CE, Garcia-Marcos M. Smooth operator(s): dialing up and down neurotransmitter responses by G-protein regulators. Trends Cell Biol 2024:S0962-8924(24)00140-5. [PMID: 39054106 DOI: 10.1016/j.tcb.2024.07.002] [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: 05/09/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
G-protein-coupled receptors (GPCRs) are essential mediators of neuromodulation and prominent pharmacological targets. While activation of heterotrimeric G-proteins (Gαβɣ) by GPCRs is essential in this process, much less is known about the postreceptor mechanisms that influence G-protein activity. Neurons express G-protein regulators that shape the amplitude and kinetics of GPCR-mediated synaptic responses. Although many of these operate by directly altering how G-proteins handle guanine-nucleotides enzymatically, recent discoveries have revealed alternative mechanisms by which GPCR-stimulated G-protein responses are modulated at the synapse. In this review, we cover the molecular basis for, and consequences of, the action of two G-protein regulators that do not affect the enzymatic activity of G-proteins directly: Gα inhibitory interacting protein (GINIP), which binds active Gα subunits, and potassium channel tetramerization domain-containing 12 (KCTD12), which binds active Gβγ subunits.
Collapse
Affiliation(s)
- Clementine E Philibert
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Mikel Garcia-Marcos
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; Department of Biology, College of Arts and Sciences, Boston University, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Qiao LY. Satellite Glial Cells Bridge Sensory Neuron Crosstalk in Visceral Pain and Cross-Organ Sensitization. J Pharmacol Exp Ther 2024; 390:213-221. [PMID: 38777604 PMCID: PMC11264254 DOI: 10.1124/jpet.123.002061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Following colonic inflammation, the uninjured bladder afferent neurons are also activated. The mechanisms and pathways underlying this sensory neuron cross-activation (from injured neurons to uninjured neurons) are not fully understood. Colonic and bladder afferent neurons reside in the same spinal segments and are separated by satellite glial cells (SGCs) and extracellular matrix in dorsal root ganglia (DRG). SGCs communicate with sensory neurons in a bidirectional fashion. This review summarizes the differentially regulated genes/proteins in the injured and uninjured DRG neurons and explores the role of SGCs in regulation of sensory neuron crosstalk in visceral cross-organ sensitization. The review also highlights the paracrine pathways in mediating neuron-SGC and SGC-neuron coupling with an emphasis on the neurotrophins and purinergic systems. Finally, I discuss the results from recent RNAseq profiling of SGCs to reveal useful molecular markers for characterization, functional study, and therapeutic targets of SGCs. SIGNIFICANCE STATEMENT: Satellite glial cells (SGCs) are the largest glial subtypes in sensory ganglia and play a critical role in mediating sensory neuron crosstalk, an underlying mechanism in colon-bladder cross-sensitization. Identification of novel and unique molecular markers of SGCs can advance the discovery of therapeutic targets in treatment of chronic pain including visceral pain comorbidity.
Collapse
Affiliation(s)
- Liya Y Qiao
- Department of Physiology and Biophysics, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
8
|
Noe-Steinmüller N, Scherbakov D, Zhuravlyova A, Wager TD, Goldstein P, Tesarz J. Defining suffering in pain: a systematic review on pain-related suffering using natural language processing. Pain 2024; 165:1434-1449. [PMID: 38452202 PMCID: PMC11190900 DOI: 10.1097/j.pain.0000000000003195] [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: 07/21/2023] [Revised: 10/26/2023] [Accepted: 11/26/2023] [Indexed: 03/09/2024]
Abstract
ABSTRACT Understanding, measuring, and mitigating pain-related suffering is a key challenge for both clinical care and pain research. However, there is no consensus on what exactly the concept of pain-related suffering includes, and it is often not precisely operationalized in empirical studies. Here, we (1) systematically review the conceptualization of pain-related suffering in the existing literature, (2) develop a definition and a conceptual framework, and (3) use machine learning to cross-validate the results. We identified 111 articles in a systematic search of Web of Science, PubMed, PsychINFO, and PhilPapers for peer-reviewed articles containing conceptual contributions about the experience of pain-related suffering. We developed a new procedure for extracting and synthesizing study information based on the cross-validation of qualitative analysis with an artificial intelligence-based approach grounded in large language models and topic modeling. We derived a definition from the literature that is representative of current theoretical views and describes pain-related suffering as a severely negative, complex, and dynamic experience in response to a perceived threat to an individual's integrity as a self and identity as a person. We also offer a conceptual framework of pain-related suffering distinguishing 8 dimensions: social, physical, personal, spiritual, existential, cultural, cognitive, and affective. Our data show that pain-related suffering is a multidimensional phenomenon that is closely related to but distinct from pain itself. The present analysis provides a roadmap for further theoretical and empirical development.
Collapse
Affiliation(s)
- Niklas Noe-Steinmüller
- Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | | | | | - Jonas Tesarz
- Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
9
|
Qarot E, Guan Y, Hanani M. The protective barrier role of satellite glial cells in sensory ganglia. Glia 2024; 72:1054-1066. [PMID: 38450799 DOI: 10.1002/glia.24511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/11/2024] [Accepted: 01/24/2024] [Indexed: 03/08/2024]
Abstract
Neurons in sensory ganglia are wrapped completely by satellite glial cells (SGCs). One putative function of SGCs is to regulate the neuronal microenvironment, but this role has received only little attention. In this study we investigated whether the SGC envelope serves a barrier function and how SGCs may control the neuronal microenvironment. We studied this question on short-term (<24 h) cell cultures of dorsal root ganglia and trigeminal ganglia from adult mice, which contain neurons surrounded with SGCs, and neurons that are not. Using calcium imaging, we measured neuronal responses to molecules with established actions on sensory neurons. We found that neurons surrounded by SGCs had a smaller response to molecules such as adenosine triphosphate (ATP), glutamate, GABA, and bradykinin than neurons without glial cover. When we inhibited the activity of NTPDases, which hydrolyze the ATP, and also when we inhibited the glutamate and GABA transporters on SGCs, this difference in the neuronal response was no longer observed. We conclude that the SGC envelope does not hinder diffusional passage, but acts as a metabolic barrier that regulates the neuronal microenvironment, and can protect the neurons and modulate their activity.
Collapse
Affiliation(s)
- Eman Qarot
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
- Department of Neurological Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
| | - Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
10
|
Stanton E, Won P, Manasyan A, Gurram S, Gilllenwater TJ, Yenikomshian HA. Neuropathic pain in burn patients - A common problem with little literature: A systematic review. Burns 2024; 50:1053-1061. [PMID: 38472004 PMCID: PMC11216128 DOI: 10.1016/j.burns.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/27/2023] [Accepted: 02/21/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND The prevalence of neuropathic pain (NP) in burn patients is reported in the literature to be as high as 80%1. Given the complexity of NP in burn patients and the wide range of treatments available, a systematic review of the literature is warranted to summarize our current understanding of management and treatment of NP in this population. METHODS This systematic review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. The following databases were queried to identify relevant articles: PubMed, Cochrane, Embase, Scopus, Ovid, and Web of Science. The main outcome measures were incidence and management of NP. Secondary outcomes included risk factors for NP. RESULTS Included articles presented findings from 11 different countries, capturing outcomes for 4366 patients. Risk factors for neuropathic pain in burn patients were identified, including older age, alcohol and substance abuse, current daily smoking, greater % total body surface area burns (TBSA), and longer hospitalizations. Pharmacologic treatments included gabapentin/pregabalin (n = 7), ascorbic acid (n = 1), and lidocaine (n = 1). Overall, the studies showed varied results regarding the efficacy of pharmacological treatments. While certain studies demonstrated gabapentanoids to be effective in reducing neuropathic symptoms, others found conflicting results. With regards to non-pharmacologic treatments, electroconvulsive therapy (n = 1), electropuncture (n = 1), nerve release/reconstruction (n = 2), and somatosensory feedback rehabilitation (n = 1) were used and demonstrated promise in reducing pain intensity and improving functionality. CONCLUSIONS Despite NP afflicting the majority of burn patients long after their injury, this systematic review demonstrates insufficient evidence on the pathophysiology, outcomes, and risk factors in NP, as well as the efficacy of various therapies. Future prospective and randomized studies evaluating the etiology of these factors can substantially improve our treatment strategies. This can allow for the development of well-delineated and evidence-based protocols in NP management in hopes of improving quality of life and both psychological and physical function in burn patients.
Collapse
Affiliation(s)
- Eloise Stanton
- Keck School of Medicine of USC, Los Angeles, CA, USA; Division of Plastic and Reconstructive Surgery, Keck Medicine of USC, Los Angeles, CA, USA
| | - Paul Won
- Keck School of Medicine of USC, Los Angeles, CA, USA; Division of Plastic and Reconstructive Surgery, Keck Medicine of USC, Los Angeles, CA, USA
| | | | | | - T Justin Gilllenwater
- Keck School of Medicine of USC, Los Angeles, CA, USA; Division of Plastic and Reconstructive Surgery, Keck Medicine of USC, Los Angeles, CA, USA
| | - Haig A Yenikomshian
- Keck School of Medicine of USC, Los Angeles, CA, USA; Division of Plastic and Reconstructive Surgery, Keck Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
11
|
Li WW, Zhao Y, Liu HC, Liu J, Chan SO, Zhong YF, Zhang TY, Liu Y, Zhang W, Xia YQ, Chi XC, Xu J, Wang Y, Wang J. Roles of Thermosensitive Transient Receptor Channels TRPV1 and TRPM8 in Paclitaxel-Induced Peripheral Neuropathic Pain. Int J Mol Sci 2024; 25:5813. [PMID: 38892000 PMCID: PMC11171746 DOI: 10.3390/ijms25115813] [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: 04/26/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Paclitaxel, a microtubule-stabilizing chemotherapy drug, can cause severe paclitaxel-induced peripheral neuropathic pain (PIPNP). The roles of transient receptor potential (TRP) ion channel vanilloid 1 (TRPV1, a nociceptor and heat sensor) and melastatin 8 (TRPM8, a cold sensor) in PIPNP remain controversial. In this study, Western blotting, immunofluorescence staining, and calcium imaging revealed that the expression and functional activity of TRPV1 were upregulated in rat dorsal root ganglion (DRG) neurons in PIPNP. Behavioral assessments using the von Frey and brush tests demonstrated that mechanical hyperalgesia in PIPNP was significantly inhibited by intraperitoneal or intrathecal administration of the TRPV1 antagonist capsazepine, indicating that TRPV1 played a key role in PIPNP. Conversely, the expression of TRPM8 protein decreased and its channel activity was reduced in DRG neurons. Furthermore, activation of TRPM8 via topical application of menthol or intrathecal injection of WS-12 attenuated the mechanical pain. Mechanistically, the TRPV1 activity triggered by capsaicin (a TRPV1 agonist) was reduced after menthol application in cultured DRG neurons, especially in the paclitaxel-treated group. These findings showed that upregulation of TRPV1 and inhibition of TRPM8 are involved in the generation of PIPNP, and they suggested that inhibition of TRPV1 function in DRG neurons via activation of TRPM8 might underlie the analgesic effects of menthol.
Collapse
Affiliation(s)
- Wen-Wen Li
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Yan Zhao
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Huai-Cun Liu
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Jiao Liu
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing 100191, China;
| | - Sun-On Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yi-Fei Zhong
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Tang-Yu Zhang
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Yu Liu
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Wei Zhang
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Yu-Qi Xia
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Xiao-Chun Chi
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Jian Xu
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| | - Yun Wang
- Neuroscience Research Institute and Department of Neurobiology, Key Laboratory for Neuroscience of Ministry of Education and Neuroscience, Peking University Health Science Center, Beijing 100191, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Jun Wang
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (W.-W.L.); (Y.Z.); (H.-C.L.); (Y.-F.Z.); (T.-Y.Z.); (Y.L.); (W.Z.); (Y.-Q.X.); (X.-C.C.); (J.X.)
| |
Collapse
|
12
|
Shin SM, Itson-Zoske B, Fan F, Xiao Y, Qiu C, Cummins TR, Hogan QH, Yu H. Peripherally targeted analgesia via AAV-mediated sensory neuron-specific inhibition of multiple pronociceptive sodium channels. J Clin Invest 2024; 134:e170813. [PMID: 38722683 PMCID: PMC11213509 DOI: 10.1172/jci170813] [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: 03/23/2023] [Accepted: 05/07/2024] [Indexed: 06/30/2024] Open
Abstract
This study reports that targeting intrinsically disordered regions of the voltage-gated sodium channel 1.7 (NaV1.7) protein facilitates discovery of sodium channel inhibitory peptide aptamers (NaViPA) for adeno-associated virus-mediated (AAV-mediated), sensory neuron-specific analgesia. A multipronged inhibition of INa1.7, INa1.6, INa1.3, and INa1.1 - but not INa1.5 and INa1.8 - was found for a prototype and named NaViPA1, which was derived from the NaV1.7 intracellular loop 1, and is conserved among the TTXs NaV subtypes. NaViPA1 expression in primary sensory neurons (PSNs) of dorsal root ganglia (DRG) produced significant inhibition of TTXs INa but not TTXr INa. DRG injection of AAV6-encoded NaViPA1 significantly attenuated evoked and spontaneous pain behaviors in both male and female rats with neuropathic pain induced by tibial nerve injury (TNI). Whole-cell current clamp of the PSNs showed that NaViPA1 expression normalized PSN excitability in TNI rats, suggesting that NaViPA1 attenuated pain by reversal of injury-induced neuronal hypersensitivity. IHC revealed efficient NaViPA1 expression restricted in PSNs and their central and peripheral terminals, indicating PSN-restricted AAV biodistribution. Inhibition of sodium channels by NaViPA1 was replicated in the human iPSC-derived sensory neurons. These results summate that NaViPA1 is a promising analgesic lead that, combined with AAV-mediated PSN-specific block of multiple TTXs NaVs, has potential as a peripheral nerve-restricted analgesic therapeutic.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Fan Fan
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yucheng Xiao
- Department of Biology, School of Science, Indiana University-Purdue University, Indianapolis, Indiana, USA
| | - Chensheng Qiu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao, China
| | - Theodore R. Cummins
- Department of Biology, School of Science, Indiana University-Purdue University, Indianapolis, Indiana, USA
| | - Quinn H. Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
13
|
Konnova EA, Deftu AF, Chu Sin Chung P, Kirschmann G, Decosterd I, Suter MR. Potassium channel modulation in macrophages sensitizes dorsal root ganglion neurons after nerve injury. Glia 2024; 72:677-691. [PMID: 38108588 DOI: 10.1002/glia.24496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/19/2023]
Abstract
Macrophages and satellite glial cells are found between injured and uninjured neurons in the lumbar dorsal root ganglia (DRG). We explored the mechanism of neuro-immune and neuron-glia crosstalk leading to hyperexcitability of DRG neurons. After spared nerve injury (SNI), CX3CR1+ resident macrophages became activated, proliferated, and increased inward-rectifying potassium channel Kir 2.1 currents. Conditioned medium (CM) by macrophages, obtained from DRG of SNI mice, sensitized small DRG neurons from naïve mice. However, treatment with CM from GFAP+ glial cells did not affect neuronal excitability. When subjected to this macrophage-derived CM, DRG neurons had increased spontaneous activity, current-evoked responses and voltage-gated NaV 1.7 and NaV 1.8 currents. Silencing Kir 2.1 in macrophages after SNI prevented the induction of neuronal hyperexcitability from their CM. Blocking vesicular exocytosis or soluble tumor necrosis factor in CM or interfering with the downstream intracellular p38 pathway in neurons, also prevented neuronal hyperexcitability. Blocking protein trafficking in neurons reduced the effect of CM, suggesting that the hyperexcitable state resulted from changes in NaV channel trafficking. These results suggest that DRG macrophages, primed by peripheral nerve injury, contribute to neuron-glia crosstalk, NaV channel dysregulation and neuronal hyperexcitability implicated in the development of neuropathic pain.
Collapse
Affiliation(s)
- Elena A Konnova
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Alexandru-Florian Deftu
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Paul Chu Sin Chung
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Guylène Kirschmann
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Isabelle Decosterd
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Marc R Suter
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
14
|
Hanani M. Satellite Glial Cells in Human Disease. Cells 2024; 13:566. [PMID: 38607005 PMCID: PMC11011452 DOI: 10.3390/cells13070566] [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: 02/26/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
Satellite glial cells (SGCs) are the main type of glial cells in sensory ganglia. Animal studies have shown that these cells play essential roles in both normal and disease states. In a large number of pain models, SGCs were activated and contributed to the pain behavior. Much less is known about SGCs in humans, but there is emerging recognition that SGCs in humans are altered in a variety of clinical states. The available data show that human SGCs share some essential features with SGCs in rodents, but many differences do exist. SGCs in DRG from patients suffering from common painful diseases, such as rheumatoid arthritis and fibromyalgia, may contribute to the pain phenotype. It was found that immunoglobulins G (IgG) from fibromyalgia patients can induce pain-like behavior in mice. Moreover, these IgGs bind preferentially to SGCs and activate them, which can sensitize the sensory neurons, causing nociception. In other human diseases, the evidence is not as direct as in fibromyalgia, but it has been found that an antibody from a patient with rheumatoid arthritis binds to mouse SGCs, which leads to the release of pronociceptive factors from them. Herpes zoster is another painful disease, and it appears that the zoster virus resides in SGCs, which acquire an abnormal morphology and may participate in the infection and pain generation. More work needs to be undertaken on SGCs in humans, and this review points to several promising avenues for better understanding disease mechanisms and developing effective pain therapies.
Collapse
Affiliation(s)
- Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem 91240, Israel; ; Tel.: +972-2-5844721
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| |
Collapse
|
15
|
Viudez-Martínez A, Torregrosa AB, Navarrete F, García-Gutiérrez MS. Understanding the Biological Relationship between Migraine and Depression. Biomolecules 2024; 14:163. [PMID: 38397400 PMCID: PMC10886628 DOI: 10.3390/biom14020163] [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: 12/27/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
Migraine is a highly prevalent neurological disorder. Among the risk factors identified, psychiatric comorbidities, such as depression, seem to play an important role in its onset and clinical course. Patients with migraine are 2.5 times more likely to develop a depressive disorder; this risk becomes even higher in patients suffering from chronic migraine or migraine with aura. This relationship is bidirectional, since depression also predicts an earlier/worse onset of migraine, increasing the risk of migraine chronicity and, consequently, requiring a higher healthcare expenditure compared to migraine alone. All these data suggest that migraine and depression may share overlapping biological mechanisms. Herein, this review explores this topic in further detail: firstly, by introducing the common epidemiological and risk factors for this comorbidity; secondly, by focusing on providing the cumulative evidence of common biological aspects, with a particular emphasis on the serotoninergic system, neuropeptides such as calcitonin-gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP), substance P, neuropeptide Y and orexins, sexual hormones, and the immune system; lastly, by remarking on the future challenges required to elucidate the etiopathological mechanisms of migraine and depression and providing updated information regarding new key targets for the pharmacological treatment of these clinical entities.
Collapse
Affiliation(s)
- Adrián Viudez-Martínez
- Hospital Pharmacy Service, Hospital General Dr. Balmis de Alicante, 03010 Alicante, Spain;
| | - Abraham B. Torregrosa
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
16
|
Gadepalli A, Ummadisetty O, Akhilesh, Chouhan D, Yadav KE, Tiwari V. Peripheral mu-opioid receptor activation by dermorphin [D-Arg2, Lys4] (1-4) amide alleviates behavioral and neurobiological aberrations in rat model of chemotherapy-induced neuropathic pain. Neurotherapeutics 2024; 21:e00302. [PMID: 38241153 PMCID: PMC10903092 DOI: 10.1016/j.neurot.2023.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 01/21/2024] Open
Abstract
Paclitaxel, a frequently utilized chemotherapeutic agent, often gives rise to severe and distressing sensory neuropathy in patients undergoing chemotherapy. Unfortunately, current therapeutics for chemotherapy-induced neuropathic pain (CINP) demonstrate limited effectiveness and are burdened with the potential for central side effects such as sedation, respiratory depression, cognitive impairment, and addiction, posing substantial clinical challenges. In light of these limitations, present study is designed to investigate the therapeutic potential of Dermorphin [D-Arg2, Lys4] (1-4) amide (DALDA), a preferential peripherally acting mu-opioid receptor agonist, in rat model of CINP. The primary objective was to assess the analgesic properties of DALDA and elucidate the underlying mechanisms governing its therapeutic activity. Our findings revealed that DALDA treatment significantly ameliorated paclitaxel-induced evoked and spontaneous ongoing pain in rats without causing drug addiction and other central side effects. Molecular analyses further unveiled that paclitaxel administration resulted in increased expression of TRP channels, NR2B, voltage-gated sodium channels (VGSCs) and neuroinflammatory markers in both the dorsal root ganglion (DRG) and the spinal cord (L4-L5 region) of rats. DALDA treatment significantly downregulated ion channels (TRPs, VGSCs) and NR2B expressions, concomitant with the inhibition of microglial activation, resulting in the suppression of oxido-nitrosative stress and neuroinflammatory cascade. Findings from the current study suggests that peripheral mu-opioid receptors may offer a potential target for the treatment of patients suffering from CINP, offering new avenues for improved pain relief while minimizing central side effects.
Collapse
Affiliation(s)
- Anagha Gadepalli
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Obulapathi Ummadisetty
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Krushna Eknath Yadav
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
17
|
Uniyal A, Tiwari V, Tsukamoto T, Dong X, Guan Y, Raja SN. Targeting sensory neuron GPCRs for peripheral neuropathic pain. Trends Pharmacol Sci 2023; 44:1009-1027. [PMID: 37977131 PMCID: PMC10657387 DOI: 10.1016/j.tips.2023.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 11/19/2023]
Abstract
Despite the high prevalence of peripheral neuropathic pain (NP) conditions and significant progress in understanding its underlying mechanisms, the management of peripheral NP remains inadequate. Existing pharmacotherapies for NP act primarily on the central nervous system (CNS) and are often associated with CNS-related adverse effects, limiting their clinical effectiveness. Mounting preclinical evidence indicates that reducing the heightened activity in primary sensory neurons by targeting G-protein-coupled receptors (GPCRs), without activating these receptors in the CNS, relieves pain without central adverse effects. In this review, we focus on recent advancements in GPCR-mediated peripheral pain relief and discuss strategies to advance the development of more effective and safer therapies for peripheral NP by shifting from traditional CNS modulatory approaches toward selective targeting of GPCRs on primary sensory neurons.
Collapse
Affiliation(s)
- Ankit Uniyal
- Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (B.H.U), Varanasi, India
| | - Takashi Tsukamoto
- Department of Neurology and Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xinzhong Dong
- Department of Neuroscience, The Johns Hopkins University, Baltimore, MD, USA
| | - Yun Guan
- Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA; Department of Neurological Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Srinivasa N Raja
- Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA; Department of Neurology and Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
18
|
Tibbs GR, Uprety R, Warren JD, Beyer NP, Joyce RL, Ferrer MA, Mellado W, Wong VSC, Goldberg DC, Cohen MW, Costa CJ, Li Z, Zhang G, Dephoure NE, Barman DN, Sun D, Ingólfsson HI, Sauve AA, Willis DE, Goldstein PA. An anchor-tether 'hindered' HCN1 inhibitor is antihyperalgesic in a rat spared nerve injury neuropathic pain model. Br J Anaesth 2023; 131:745-763. [PMID: 37567808 PMCID: PMC10541997 DOI: 10.1016/j.bja.2023.06.067] [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: 04/13/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Neuropathic pain impairs quality of life, is widely prevalent, and incurs significant costs. Current pharmacological therapies have poor/no efficacy and significant adverse effects; safe and effective alternatives are needed. Hyperpolarisation-activated cyclic nucleotide-regulated (HCN) channels are causally implicated in some forms of peripherally mediated neuropathic pain. Whilst 2,6-substituted phenols, such as 2,6-di-tert-butylphenol (26DTB-P), selectively inhibit HCN1 gating and are antihyperalgesic, the development of therapeutically tolerable, HCN-selective antihyperalgesics based on their inverse agonist activity requires that such drugs spare the cardiac isoforms and do not cross the blood-brain barrier. METHODS In silico molecular dynamics simulation, in vitro electrophysiology, and in vivo rat spared nerve injury methods were used to test whether 'hindered' variants of 26DTB-P (wherein a hydrophilic 'anchor' is attached in the para-position of 26DTB-P via an acyl chain 'tether') had the desired properties. RESULTS Molecular dynamics simulation showed that membrane penetration of hindered 26DTB-Ps is controlled by a tethered diol anchor without elimination of head group rotational freedom. In vitro and in vivo analysis showed that BP4L-18:1:1, a variant wherein a diol anchor is attached to 26DTB-P via an 18-carbon tether, is an HCN1 inverse agonist and an orally available antihyperalgesic. With a CNS multiparameter optimisation score of 2.25, a >100-fold lower drug load in the brain vs blood, and an absence of adverse cardiovascular or CNS effects, BP4L-18:1:1 was shown to be poorly CNS penetrant and cardiac sparing. CONCLUSIONS These findings provide a proof-of-concept demonstration that anchor-tethered drugs are a new chemotype for treatment of disorders involving membrane targets.
Collapse
Affiliation(s)
- Gareth R Tibbs
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Rajendra Uprety
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - J David Warren
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Nicole P Beyer
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Rebecca L Joyce
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Matthew A Ferrer
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | | | | | | | | | | | - Zhucui Li
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Guoan Zhang
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Noah E Dephoure
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Dipti N Barman
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Delin Sun
- Lawrence Livermore National Laboratory, Livermore, CA, USA
| | | | - Anthony A Sauve
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Dianna E Willis
- Burke Neurological Institute, White Plains, NY, USA; Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Peter A Goldstein
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA; Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
19
|
Yang X, Chen C, Wang K, Chen M, Wang Y, Chen Z, Zhao W, Ou S. Elucidating the molecular mechanisms of ozone therapy for neuropathic pain management by integrated transcriptomic and metabolomic approach. Front Genet 2023; 14:1231682. [PMID: 37779912 PMCID: PMC10536237 DOI: 10.3389/fgene.2023.1231682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/18/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction: Neuropathic pain remains a prevalent and challenging condition to treat, with current therapies often providing inadequate relief. Ozone therapy has emerged as a promising treatment option; however, its mechanisms of action in neuropathic pain remain poorly understood. Methods: In this study, we investigated the effects of ozone treatment on gene expression and metabolite levels in the brainstem and hypothalamus of a rat model, using a combined transcriptomic and metabolomic approach. Results: Our findings revealed significant alterations in key genes, including DCST1 and AIF1L, and metabolites such as Aconitic acid, L-Glutamic acid, UDP-glucose, and Tyrosine. These changes suggest a complex interplay of molecular pathways and region-specific mechanisms underlying the analgesic effects of ozone therapy. Discussion: Our study provides insights into the molecular targets of ozone treatment for neuropathic pain, laying the groundwork for future research on validating these targets and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Xiaolan Yang
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chaoming Chen
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Keyang Wang
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Chen
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Wang
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengping Chen
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wang Zhao
- Department of Neurology, Yongchuan Hospital Affiliated to Chongqing Medical University, Chongqing, China
| | - Shu Ou
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
20
|
Liu H, Lauzadis J, Gunaratna K, Sipple E, Kaczocha M, Puopolo M. Inhibition of T-Type Calcium Channels With TTA-P2 Reduces Chronic Neuropathic Pain Following Spinal Cord Injury in Rats. THE JOURNAL OF PAIN 2023; 24:1681-1695. [PMID: 37169156 DOI: 10.1016/j.jpain.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/03/2023] [Accepted: 05/02/2023] [Indexed: 05/13/2023]
Abstract
Spinal cord injury (SCI)-induced neuropathic pain (SCI-NP) develops in up to 60 to 70% of people affected by traumatic SCI, leading to a major decline in quality of life and increased risk for depression, anxiety, and addiction. Gabapentin and pregabalin, together with antidepressant drugs, are commonly prescribed to treat SCI-NP, but their efficacy is unsatisfactory. The limited efficacy of current pharmacological treatments for SCI-NP likely reflects our limited knowledge of the underlying mechanism(s) responsible for driving the maintenance of SCI-NP. The leading hypothesis in the field supports a major role for spontaneously active injured nociceptors in driving the maintenance of SCI-NP. Recent data from our laboratory provided additional support for this hypothesis and identified the T-type calcium channels as key players in driving the spontaneous activity of SCI-nociceptors, thus providing a rational pharmacological target to treat SCI-NP. To test whether T-type calcium channels contribute to the maintenance of SCI-NP, male and female SCI and sham rats were treated with TTA-P2 (a blocker of T-type calcium channels) to determine its effects on mechanical hypersensitivity (as measured with the von Frey filaments) and spontaneous ongoing pain (as measured with the conditioned place preference paradigm), and compared them to the effects of gabapentin, a blocker of high voltage-activated calcium channels. We found that both TTA-P2 and gabapentin reduced mechanical hypersensitivity in male and females SCI rats, but surprisingly only TTA-P2 reduced spontaneous ongoing pain in male SCI rats. PERSPECTIVES: SCI-induced neuropathic pain, and in particular the spontaneous ongoing pain component, is notoriously very difficult to treat. Our data provide evidence that inhibition of T-type calcium channels reduces spontaneous ongoing pain in SCI rats, supporting a clinically relevant role for T-type channels in the maintenance of SCI-induced neuropathic pain.
Collapse
Affiliation(s)
- Huilin Liu
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Justas Lauzadis
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Kavindu Gunaratna
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Erin Sipple
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York.
| |
Collapse
|
21
|
Szewczyk AK, Ulutas S, Aktürk T, Al-Hassany L, Börner C, Cernigliaro F, Kodounis M, Lo Cascio S, Mikolajek D, Onan D, Ragaglini C, Ratti S, Rivera-Mancilla E, Tsanoula S, Villino R, Messlinger K, Maassen Van Den Brink A, de Vries T. Prolactin and oxytocin: potential targets for migraine treatment. J Headache Pain 2023; 24:31. [PMID: 36967387 PMCID: PMC10041814 DOI: 10.1186/s10194-023-01557-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/28/2023] [Indexed: 03/28/2023] Open
Abstract
Migraine is a severe neurovascular disorder of which the pathophysiology is not yet fully understood. Besides the role of inflammatory mediators that interact with the trigeminovascular system, cyclic fluctuations in sex steroid hormones are involved in the sex dimorphism of migraine attacks. In addition, the pituitary-derived hormone prolactin and the hypothalamic neuropeptide oxytocin have been reported to play a modulating role in migraine and contribute to its sex-dependent differences. The current narrative review explores the relationship between these two hormones and the pathophysiology of migraine. We describe the physiological role of prolactin and oxytocin, its relationship to migraine and pain, and potential therapies targeting these hormones or their receptors.In summary, oxytocin and prolactin are involved in nociception in opposite ways. Both operate at peripheral and central levels, however, prolactin has a pronociceptive effect, while oxytocin appears to have an antinociceptive effect. Therefore, migraine treatment targeting prolactin should aim to block its effects using prolactin receptor antagonists or monoclonal antibodies specifically acting at migraine-pain related structures. This action should be local in order to avoid a decrease in prolactin levels throughout the body and associated adverse effects. In contrast, treatment targeting oxytocin should enhance its signalling and antinociceptive effects, for example using intranasal administration of oxytocin, or possibly other oxytocin receptor agonists. Interestingly, the prolactin receptor and oxytocin receptor are co-localized with estrogen receptors as well as calcitonin gene-related peptide and its receptor, providing a positive perspective on the possibilities for an adequate pharmacological treatment of these nociceptive pathways. Nevertheless, many questions remain to be answered. More particularly, there is insufficient data on the role of sex hormones in men and the correct dosing according to sex differences, hormonal changes and comorbidities. The above remains a major challenge for future development.
Collapse
Affiliation(s)
- Anna K Szewczyk
- Doctoral School, Medical University of Lublin, Lublin, Poland
- Department of Neurology, Medical University of Lublin, Lublin, Poland
| | - Samiye Ulutas
- Department of Neurology, Kartal Dr. Lutfi Kirdar Research and Training Hospital, Istanbul, Turkey
| | - Tülin Aktürk
- Department of Neurology, Kartal Dr. Lutfi Kirdar Research and Training Hospital, Istanbul, Turkey
| | - Linda Al-Hassany
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Corinna Börner
- Department of Pediatrics - Dr. von Hauner Children's Hospital, LMU Hospital, Division of Pediatric Neurology and Developmental Medicine, Ludwig-Maximilians Universität München, Lindwurmstr. 4, 80337, Munich, Germany
- LMU Center for Children with Medical Complexity - iSPZ Hauner, Ludwig-Maximilians-Universität München, Lindwurmstr. 4, 80337, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Federica Cernigliaro
- Child Neuropsychiatry Unit Department, Pro.M.I.S.E. "G D'Alessandro, University of Palermo, 90133, Palermo, Italy
| | - Michalis Kodounis
- First Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Salvatore Lo Cascio
- Child Neuropsychiatry Unit Department, Pro.M.I.S.E. "G D'Alessandro, University of Palermo, 90133, Palermo, Italy
| | - David Mikolajek
- Department of Neurology, City Hospital Ostrava, Ostrava, Czech Republic
| | - Dilara Onan
- Spine Health Unit, Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Chiara Ragaglini
- Neuroscience Section, Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, 67100, L'Aquila, Italy
| | - Susanna Ratti
- Neuroscience Section, Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, 67100, L'Aquila, Italy
| | - Eduardo Rivera-Mancilla
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sofia Tsanoula
- Department of Neurology, 401 Military Hospital of Athens, Athens, Greece
| | - Rafael Villino
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Karl Messlinger
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Antoinette Maassen Van Den Brink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tessa de Vries
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
22
|
Shin SM, Lauzadis J, Itson-Zoske B, Cai Y, Fan F, Natarajan GK, Kwok WM, Puopolo M, Hogan QH, Yu H. Targeting intrinsically disordered regions facilitates discovery of calcium channels 3.2 inhibitory peptides for adeno-associated virus-mediated peripheral analgesia. Pain 2022; 163:2466-2484. [PMID: 35420557 PMCID: PMC9562599 DOI: 10.1097/j.pain.0000000000002650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/19/2022] [Accepted: 03/23/2022] [Indexed: 11/27/2022]
Abstract
ABSTRACT Ample data support a prominent role of peripheral T-type calcium channels 3.2 (Ca V 3.2) in generating pain states. Development of primary sensory neuron-specific inhibitors of Ca V 3.2 channels is an opportunity for achieving effective analgesic therapeutics, but success has been elusive. Small peptides, especially those derived from natural proteins as inhibitory peptide aptamers (iPAs), can produce highly effective and selective blockade of specific nociceptive molecular pathways to reduce pain with minimal off-target effects. In this study, we report the engineering of the potent and selective iPAs of Ca V 3.2 from the intrinsically disordered regions (IDRs) of Ca V 3.2 intracellular segments. Using established prediction algorithms, we localized the IDRs in Ca V 3.2 protein and identified several Ca V 3.2iPA candidates that significantly reduced Ca V 3.2 current in HEK293 cells stably expressing human wide-type Ca V 3.2. Two prototype Ca V 3.2iPAs (iPA1 and iPA2) derived from the IDRs of Ca V 3.2 intracellular loops 2 and 3, respectively, were expressed selectively in the primary sensory neurons of dorsal root ganglia in vivo using recombinant adeno-associated virus (AAV), which produced sustained inhibition of calcium current conducted by Ca V 3.2/T-type channels and significantly attenuated both evoked and spontaneous pain behavior in rats with neuropathic pain after tibial nerve injury. Recordings from dissociated sensory neurons showed that AAV-mediated Ca V 3.2iPA expression suppressed neuronal excitability, suggesting that Ca V 3.2iPA treatment attenuated pain by reversal of injury-induced neuronal hypersensitivity. Collectively, our results indicate that Ca V 3.2iPAs are promising analgesic leads that, combined with AAV-mediated delivery in anatomically targeted sensory ganglia, have the potential to be a selective peripheral Ca V 3.2-targeting strategy for clinical treatment of pain.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Justas Lauzadis
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Yongsong Cai
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Fan Fan
- Department of Pharmacology and Toxicology, The University of Mississippi Medical Center, Jackson, MS, United States
| | - Gayathri K. Natarajan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| | - Quinn H. Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
23
|
North RY, Odem MA, Li Y, Tatsui CE, Cassidy RM, Dougherty PM, Walters ET. Electrophysiological Alterations Driving Pain-Associated Spontaneous Activity in Human Sensory Neuron Somata Parallel Alterations Described in Spontaneously Active Rodent Nociceptors. THE JOURNAL OF PAIN 2022; 23:1343-1357. [PMID: 35292377 PMCID: PMC9357108 DOI: 10.1016/j.jpain.2022.02.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 06/10/2023]
Abstract
Neuropathic pain in rodents can be driven by ectopic spontaneous activity (SA) generated by sensory neurons in dorsal root ganglia (DRG). The recent demonstration that SA in dissociated human DRG neurons is associated with reported neuropathic pain in patients enables a detailed comparison of pain-linked electrophysiological alterations driving SA in human DRG neurons to alterations that distinguish SA in nociceptors from SA in low-threshold mechanoreceptors (LTMRs) in rodent neuropathy models. Analysis of recordings from dissociated somata of patient-derived DRG neurons showed that SA and corresponding pain in both sexes were significantly associated with the three functional electrophysiological alterations sufficient to generate SA in the absence of extrinsic depolarizing inputs. These include enhancement of depolarizing spontaneous fluctuations of membrane potential (DSFs), which were analyzed quantitatively for the first time in human DRG neurons. The functional alterations were indistinguishable from SA-driving alterations reported for nociceptors in rodent chronic pain models. Irregular, low-frequency DSFs in human DRG neurons closely resemble DSFs described in rodent nociceptors while differing substantially from the high-frequency sinusoidal oscillations described in rodent LTMRs. These findings suggest that conserved physiological mechanisms of SA in human nociceptor somata can drive neuropathic pain despite documented cellular differences between human and rodent DRG neurons. PERSPECTIVE: Electrophysiological alterations in human sensory neurons associated with patient-reported neuropathic pain include all three of the functional alterations that logically can promote spontaneous activity. The similarity of distinctively altered spontaneous depolarizations in human DRG neurons and rodent nociceptors suggests that spontaneously active human nociceptors can persistently promote neuropathic pain in patients.
Collapse
Affiliation(s)
- Robert Y North
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Max A Odem
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth, Houston, Texas
| | - Yan Li
- Department of Anesthesia and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Claudio Esteves Tatsui
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ryan M Cassidy
- M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Patrick M Dougherty
- Department of Anesthesia and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas..
| |
Collapse
|
24
|
Mechanisms of bone pain: Progress in research from bench to bedside. Bone Res 2022; 10:44. [PMID: 35668080 PMCID: PMC9170780 DOI: 10.1038/s41413-022-00217-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 12/27/2022] Open
Abstract
AbstractThe field of research on pain originating from various bone diseases is expanding rapidly, with new mechanisms and targets asserting both peripheral and central sites of action. The scope of research is broadening from bone biology to neuroscience, neuroendocrinology, and immunology. In particular, the roles of primary sensory neurons and non-neuronal cells in the peripheral tissues as important targets for bone pain treatment are under extensive investigation in both pre-clinical and clinical settings. An understanding of the peripheral mechanisms underlying pain conditions associated with various bone diseases will aid in the appropriate application and development of optimal strategies for not only managing bone pain symptoms but also improving bone repairing and remodeling, which potentially cures the underlying etiology for long-term functional recovery. In this review, we focus on advances in important preclinical studies of significant bone pain conditions in the past 5 years that indicated new peripheral neuronal and non-neuronal mechanisms, novel targets for potential clinical interventions, and future directions of research.
Collapse
|
25
|
Carta G, Fornasari BE, Fregnan F, Ronchi G, De Zanet S, Muratori L, Nato G, Fogli M, Gambarotta G, Geuna S, Raimondo S. Neurodynamic Treatment Promotes Mechanical Pain Modulation in Sensory Neurons and Nerve Regeneration in Rats. Biomedicines 2022; 10:biomedicines10061296. [PMID: 35740318 PMCID: PMC9220043 DOI: 10.3390/biomedicines10061296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 05/24/2022] [Accepted: 05/29/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Somatic nerve injuries are a rising problem leading to disability associated with neuropathic pain commonly reported as mechanical allodynia (MA) and hyperalgesia. These symptoms are strongly dependent on specific processes in the dorsal root ganglia (DRG). Neurodynamic treatment (NDT), consisting of selective uniaxial nerve repeated tension protocols, effectively reduces pain and disability in neuropathic pain patients even though the biological mechanisms remain poorly characterized. We aimed to define, both in vivo and ex vivo, how NDT could promote nerve regeneration and modulate some processes in the DRG linked to MA and hyperalgesia. Methods: We examined in Wistar rats, after unilateral median and ulnar nerve crush, the therapeutic effects of NDT and the possible protective effects of NDT administered for 10 days before the injury. We adopted an ex vivo model of DRG organotypic explant subjected to NDT to explore the selective effects on DRG cells. Results: Behavioural tests, morphological and morphometrical analyses, and gene and protein expression analyses were performed, and these tests revealed that NDT promotes nerve regeneration processes, speeds up sensory motor recovery, and modulates mechanical pain by affecting, in the DRG, the expression of TACAN, a mechanosensitive receptor shared between humans and rats responsible for MA and hyperalgesia. The ex vivo experiments have shown that NDT increases neurite regrowth and confirmed the modulation of TACAN. Conclusions: The results obtained in this study on the biological and molecular mechanisms induced by NDT will allow the exploration, in future clinical trials, of its efficacy in different conditions of neuropathic pain.
Collapse
Affiliation(s)
- Giacomo Carta
- Department of Clinical and Biological Sciences, University of Torino, 10043 Torino, Italy; (G.C.); (B.E.F.); (G.R.); (S.D.Z.); (L.M.); (G.G.); (S.G.); (S.R.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, 10043 Torino, Italy; (G.N.); (M.F.)
- Department of Rehabilitation, ASST (Azienda Socio Sanitaria Territoriali) Nord Milano, Sesto San Giovanni Hospital, Sesto San Giovanni, 20099 Milano, Italy
| | - Benedetta Elena Fornasari
- Department of Clinical and Biological Sciences, University of Torino, 10043 Torino, Italy; (G.C.); (B.E.F.); (G.R.); (S.D.Z.); (L.M.); (G.G.); (S.G.); (S.R.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, 10043 Torino, Italy; (G.N.); (M.F.)
| | - Federica Fregnan
- Department of Clinical and Biological Sciences, University of Torino, 10043 Torino, Italy; (G.C.); (B.E.F.); (G.R.); (S.D.Z.); (L.M.); (G.G.); (S.G.); (S.R.)
- Correspondence: ; Tel.: +39-(0)1-1670-5433; Fax: +39-(0)1-1903-8639
| | - Giulia Ronchi
- Department of Clinical and Biological Sciences, University of Torino, 10043 Torino, Italy; (G.C.); (B.E.F.); (G.R.); (S.D.Z.); (L.M.); (G.G.); (S.G.); (S.R.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, 10043 Torino, Italy; (G.N.); (M.F.)
| | - Stefano De Zanet
- Department of Clinical and Biological Sciences, University of Torino, 10043 Torino, Italy; (G.C.); (B.E.F.); (G.R.); (S.D.Z.); (L.M.); (G.G.); (S.G.); (S.R.)
| | - Luisa Muratori
- Department of Clinical and Biological Sciences, University of Torino, 10043 Torino, Italy; (G.C.); (B.E.F.); (G.R.); (S.D.Z.); (L.M.); (G.G.); (S.G.); (S.R.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, 10043 Torino, Italy; (G.N.); (M.F.)
| | - Giulia Nato
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, 10043 Torino, Italy; (G.N.); (M.F.)
- Department of Life Sciences and Systems Biology, University of Torino, 10124 Torino, Italy
| | - Marco Fogli
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, 10043 Torino, Italy; (G.N.); (M.F.)
- Department of Life Sciences and Systems Biology, University of Torino, 10124 Torino, Italy
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences, University of Torino, 10043 Torino, Italy; (G.C.); (B.E.F.); (G.R.); (S.D.Z.); (L.M.); (G.G.); (S.G.); (S.R.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, 10043 Torino, Italy; (G.N.); (M.F.)
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, University of Torino, 10043 Torino, Italy; (G.C.); (B.E.F.); (G.R.); (S.D.Z.); (L.M.); (G.G.); (S.G.); (S.R.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, 10043 Torino, Italy; (G.N.); (M.F.)
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences, University of Torino, 10043 Torino, Italy; (G.C.); (B.E.F.); (G.R.); (S.D.Z.); (L.M.); (G.G.); (S.G.); (S.R.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, 10043 Torino, Italy; (G.N.); (M.F.)
| |
Collapse
|
26
|
Exercise Reduces Pain Behavior and Pathological Changes in Dorsal Root Ganglia Induced by Systemic Inflammation in Mice. Neurosci Lett 2022; 778:136616. [DOI: 10.1016/j.neulet.2022.136616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/11/2022] [Accepted: 04/01/2022] [Indexed: 11/23/2022]
|
27
|
Satellite Glial Cells and Neurons in Trigeminal Ganglia Are Altered in an Itch Model in Mice. Cells 2022; 11:cells11050886. [PMID: 35269508 PMCID: PMC8909456 DOI: 10.3390/cells11050886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 02/04/2023] Open
Abstract
Itch (pruritus) is a common chronic condition with a lifetime prevalence of over 20%. The mechanisms underlying itch are poorly understood, and its therapy is difficult. There is recent evidence that following nerve injury or inflammation, intercellular communications in sensory ganglia are augmented, which may lead to abnormal neuronal activity, and hence to pain, but there is no information whether such changes take place in an itch model. We studied changes in neurons and satellite glial cells (SGCs) in trigeminal ganglia in an itch model in mice using repeated applications of 2,4,6-trinitro-1-chlorobenzene (TNCB) to the external ear over a period of 11 days. Treated mice showed augmented scratching behavior as compared with controls during the application period and for several days afterwards. Immunostaining for the activation marker glial fibrillary acidic protein in SGCs was greater by about 35% after TNCB application, and gap junction-mediated coupling between neurons increased from about 2% to 13%. The injection of gap junction blockers reduced scratching behavior, suggesting that gap junctions contribute to itch. Calcium imaging studies showed increased responses of SGCs to the pain (and presumed itch) mediator ATP. We conclude that changes in both neurons and SGCs in sensory ganglia may play a role in itch.
Collapse
|
28
|
Roza C, Bernal L. Electrophysiological characterization of ectopic spontaneous discharge in axotomized and intact fibers upon nerve transection: a role in spontaneous pain? Pflugers Arch 2022; 474:387-396. [PMID: 35088129 DOI: 10.1007/s00424-021-02655-7] [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: 09/28/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022]
Abstract
Many patients experience positive symptoms after traumatic nerve injury. Despite the increasing number of experimental studies in models of peripheral neuropathy and the knowledge acquired, most of these patients lack an effective treatment for their chronic pain. One possible explanation might be that most of the preclinical studies focused on the development of mechanical or thermal allodynia/hyperalgesia, neglecting that most of the patients with peripheral neuropathies complain mostly about spontaneous forms of pains. Here, we summarize the aberrant electrophysiological behavior of peripheral nerve fibers recorded in experimental models, the underlying pathophysiological mechanisms, and their relationship with the symptoms reported by patients. Upon nerve section, axotomized but also intact fibers develop ectopic spontaneous activity. Most interestingly, a proportion of axotomized fibers might present receptive fields in the skin far beyond the site of damage, indicative of a functional cross talk between neuromatose and intact fibers. All these features can be linked with some of the symptoms that neuropathic patients experience. Furthermore, we spotlight the consequence of primary afferents with different patterns of spontaneous discharge on the neural code and its relationship with chronic pain states. With this article, readers will be able to understand the pathophysiological mechanisms that might underlie some of the symptoms that experience neuropathic patients, with a special focus on spontaneous pain.
Collapse
Affiliation(s)
- Carolina Roza
- Dpto. Biología de Sistemas, Edificio de Medicina Universidad de Alcalá, 28871, Alcalá de Henares, Madrid, Spain.
| | | |
Collapse
|
29
|
Stucky CL, Mikesell AR. Cutaneous pain in disorders affecting peripheral nerves. Neurosci Lett 2021; 765:136233. [PMID: 34506882 PMCID: PMC8579816 DOI: 10.1016/j.neulet.2021.136233] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 08/16/2021] [Accepted: 09/02/2021] [Indexed: 02/08/2023]
Abstract
Our ability to quickly detect and respond to harmful environmental stimuli is vital for our safety and survival. This inherent acute pain detection is a "gift" because it both protects our body from harm and allows healing of damaged tissues [1]. Damage to tissues from trauma or disease can result in distorted or amplified nociceptor signaling and sensitization of the spinal cord and brain (Central Nervous System; CNS) pathways to normal input from light touch mechanoreceptors. Together, these processes can result in nagging to unbearable chronic pain and extreme sensitivity to light skin touch (allodynia). Unlike acute protective pain, chronic pain and allodynia serve no useful purpose and can severely reduce the quality of life of an affected person. Chronic pain can arise from impairment to peripheral neurons, a phenomenon called "peripheral neuropathic pain." Peripheral neuropathic pain can be caused by many insults that directly affect peripheral sensory neurons, including mechanical trauma, metabolic imbalance (e.g., diabetes), autoimmune diseases, chemotherapeutic agents, viral infections (e.g., shingles). These insults cause "acquired" neuropathies such as small-fiber neuropathies, diabetic neuropathy, chemotherapy-induced peripheral neuropathy, and post herpetic neuralgia. Peripheral neuropathic pain can also be caused by genetic factors and result in hereditary neuropathies that include Charcot-Marie-Tooth disease, rare channelopathies and Fabry disease. Many acquired and hereditary neuropathies affect the skin, our largest organ and protector of nearly our entire body. Here we review how cutaneous nociception (pain perceived from the skin) is altered following diseases that affect peripheral nerves that innervate the skin. We provide an overview of how noxious stimuli are detected and encoded by molecular transducers on subtypes of cutaneous afferent endings and conveyed to the CNS. Next, we discuss several acquired and hereditary diseases and disorders that cause painful or insensate (lack of sensation) cutaneous peripheral neuropathies, the symptoms and percepts patients experience, and how cutaneous afferents and other peripheral cell types are altered in function in these disorders. We highlight exciting new research areas that implicate non-neuronal skin cells, particularly keratinocytes, in cutaneous nociception and peripheral neuropathies. Finally, we conclude with ideas for innovative new directions, areas of unmet need, and potential opportunities for novel cutaneous therapeutics that may avoid CNS side effects, as well as ideas for improved translation of mechanisms identified in preclinical models to patients.
Collapse
Affiliation(s)
- Cheryl L Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States.
| | - Alexander R Mikesell
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| |
Collapse
|
30
|
Shahiri TS, Richebé P, Richard-Lalonde M, Gélinas C. Description of the validity of the Analgesia Nociception Index (ANI) and Nociception Level Index (NOL) for nociception assessment in anesthetized patients undergoing surgery: a systematized review. J Clin Monit Comput 2021; 36:623-635. [PMID: 34783941 DOI: 10.1007/s10877-021-00772-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 10/17/2021] [Indexed: 11/29/2022]
Abstract
Maintaining optimum analgesia in anesthetized patients is challenging due to the inability to self-report pain or exhibit pain-related behaviours. The Analgesia Nociception Index (ANI) (based on heart rate variability [HRV]) and the Nociception Level Index (NOL) (based on HRV, photoplethysmography, skin conductance, and temperature) both include HRV and provide continuous index monitoring for nociception assessment. The research question was: "What are the validation strategies of the NOL and ANI for nociception assessment in anesthetized patients?". The objectives were to describe and analyze the validation strategies and results. A systematized review was conducted using a comprehensive search with keywords under three concepts (nociception/pain, ANI/NOL, and validity) in four databases. A quality assessment using an adapted GRADE approach for measurement tools, and a risk of bias assessment using QUADAS-2 tool were performed by two reviewers. Out of 525 results, 15 validation studies were included. Strategies included hypothesis testing, discriminative, and criterion validation. Significant changes in ANI/NOL values were found in response to nociceptive stimuli at different opioid concentrations (hypothesis testing). Higher ANI/NOL values were observed during nociceptive stimuli (discriminative). AUCs ranging from 0.83 to 0.99 were obtained to detect nociceptive stimuli (criterion). Both technologies performed superiorly in detecting nociceptive stimuli compared to individual monitoring of HR and blood pressure. Although the aforementioned validation strategies are deemed appropriate, in the absence of a gold standard, criterion validation findings should be interpreted with caution. Moreover, reliability could be examined using test-retest with consistent ANI/NOL values during a stable time-interval.
Collapse
Affiliation(s)
- T Shiva Shahiri
- Ingram School of Nursing, McGill University, Montréal, Canada.,Centre for Nursing Research and Lady Davis Institute, Jewish General Hospital - CIUSSS West-Central Montréal, Montréal, Canada
| | - Philippe Richebé
- Department of Anesthesiology and Pain Medicine, University of Montréal, Hôpital Maisonneuve-Rosemont - CIUSSS Est-de-lÏle-de-Montréal, Montréal, Canada
| | - Melissa Richard-Lalonde
- Ingram School of Nursing, McGill University, Montréal, Canada.,Centre for Nursing Research and Lady Davis Institute, Jewish General Hospital - CIUSSS West-Central Montréal, Montréal, Canada
| | - Céline Gélinas
- Ingram School of Nursing, McGill University, Montréal, Canada. .,Centre for Nursing Research and Lady Davis Institute, Jewish General Hospital - CIUSSS West-Central Montréal, Montréal, Canada.
| |
Collapse
|
31
|
Brazenor GA, Malham GM, Teddy PJ. Can Central Sensitization after injury persist as an autonomous pain generator? - A comprehensive search for evidence. PAIN MEDICINE 2021; 23:1283-1298. [PMID: 34718773 DOI: 10.1093/pm/pnab319] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/11/2021] [Accepted: 10/21/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To conduct a comprehensive search for evidence whether Central Sensitization following an injury can act as a persistent autonomous pain generator after the inducing injury has healed. METHODS We searched Medline on PubMed and the Cochrane Library, screening 3,572 abstracts, from which 937 full text articles were obtained, with 186 of these discarded as irrelevant to the question being posed. The remaining 751 articles were studied for evidence. RESULTS Fourteen publications were judged to provide weak evidence for the hypothesis of central sensitization as a persisting autonomous pain generator, but none addressed the question directly. No strong evidence for the affirmative answer was found.Sixty-two publications were judged to provide weak evidence for a negative answer, and nine judged to provide strong evidence.Unexpectedly, serious weaknesses were discovered in the literature underpinning the validity of the clinical diagnosis of Central Sensitization in man:(i) Inappropriate extrapolation, in many publications, of laboratory animal data to humans.(ii) Failure to demonstrate the absence of peripheral pain generators which might be perpetuating Central Sensitization.(iii) Many factors now shown to confound what is being measured by quantitative sensory testing, conditioned pain modulation, and Central Sensitization Inventory. CONCLUSIONS We found no evidence proving that central sensitization can persist as an autonomous pain generator after the initiating injury has healed.Our review has also shown that the evidential basis for the diagnosis of CS in individual patients is seriously in question.
Collapse
Affiliation(s)
| | | | - Peter J Teddy
- Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
32
|
Vaitkus A, Šipylaitė J. Sensory Perception in Lumbosacral Radiculopathy with Radicular Pain: Feasibility Study of Multimodal Bedside-Suitable Somatosensory Testing. Acta Med Litu 2021; 28:97-111. [PMID: 34393632 PMCID: PMC8311840 DOI: 10.15388/amed.2021.28.1.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/29/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022] Open
Abstract
Background Somatosensory testing could be useful in stratifying pain patients and improving pain treatment guidelines. Bedside-suitable techniques are searched for application in daily clinical practice. This study aimed to characterize chronic unilateral lumbosacral radiculopathy (LSR) patients with radicular pain using multimodal bedside-suitable somatosensory testing. Materials and methods We evaluated 50 chronic unilateral LSR patients with radicular pain (LSR group) and 24 controls (Control group). Sensory testing was performed using a battery of bedside sensory tests (10g monofilament, 200–400 mN brush, Lindblom rollers with controlled 25°C and 40°C temperature, and 40g neurological pin and investigator’s finger pressure). Participants had to rate their sensory perceptions on both legs at multiple test points within L3 to S2 dermatomes. Characteristics of the testing process and sensory disturbances were analyzed. Results LSR group showed sensory disturbances in 82% of patients. The Control group showed no sensory disturbances. Sensory testing took longer (p < 0.001) in the LSR group (29.3 ± 6.5 minutes per patient) than in the Control group (20.5 ± 5.2). Nine sensory phenotypes were detected in the LSR group according to individual sensory disturbances within 5 superficial tests. Conclusions The applied multimodal bedside-suitable somatosensory testing battery is suitable for sensory evaluation and characterization of LSR patients. Grouping of allied sensory phenotypes revealed some tendencies in pain intensity characteristics.
Collapse
Affiliation(s)
- Alfredas Vaitkus
- Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Jūratė Šipylaitė
- Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
33
|
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: 5.8] [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
|
34
|
Injectable Capsaicin for the Management of Pain Due to Osteoarthritis. Molecules 2021; 26:molecules26040778. [PMID: 33546181 PMCID: PMC7913147 DOI: 10.3390/molecules26040778] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 11/17/2022] Open
Abstract
Capsaicin is a potent agonist of the TRPV1 channel, a transduction channel that is highly expressed in nociceptive fibers (pain fibers) throughout the peripheral nervous system. Given the importance of TRPV1 as one of several transduction channels in nociceptive fibers, much research has been focused on the potential therapeutic benefits of using TRPV1 antagonists for the management of pain. However, an antagonist has two limitations. First, an antagonist in principle generally only affects one receptor. Secondly, most antagonists must have an ongoing presence on the receptor to have an effect. Capsaicin overcomes both liabilities by disrupting peripheral terminals of nociceptive fibers that express TRPV1, and thereby affects all of the potential means of activating that pain fiber (not just TRPV1 function). This disruptive effect is dependent on the dose and can occur within minutes. Thus, unlike a typical receptor antagonist, continued bioavailability at the level of the receptor is not necessary. By disrupting the entire terminal of the TRPV1-expressing nociceptive fiber, capsaicin blocks all the activation mechanisms within that fiber, and not just TRPV1 function. Topical capsaicin, an FDA approved treatment for neuropathic pain, addresses pain from abnormal nociceptor activity in the superficial layers of the skin. Effects after a single administration are evident over a period of weeks to months, but in time are fully reversible. This review focuses on the rationale for using capsaicin by injection for painful conditions such as osteoarthritis (OA) and provides an update on studies completed to date.
Collapse
|
35
|
Vaitkus A, Šipylaitė J. Qualitative Sensory Testing in Outcome Prediction of Transforaminal Epidural Steroid Injection for Chronic Painful Unilateral Lumbosacral Radiculopathy: Prospective Observational Study. Pain Pract 2021; 21:618-629. [PMID: 33502060 DOI: 10.1111/papr.12997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/11/2021] [Accepted: 01/15/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Transforaminal epidural steroid injection (TFESI) is widely practiced for the treatment of radicular pain. As its effectiveness is still subject to debate, a better patient selection for TFESI is necessary. We aimed to evaluate the potential of bedside-suitable qualitative sensory testing (QualST) to determine the early effectiveness of TFESI for the treatment of chronic lumbosacral radiculopathy (LSR)-related pain. METHODS Thirty-six patients with chronic painful unilateral LSR were evaluated in a prospective observational study using five standardized sensory tests (10 g monofilament, 200 to 400 mN brush, Lindblom rollers with controlled 25 and 40°C temperature, and 40 g neurological pin). Quality of sensory perceptions on the painful leg was compared to the non-painful leg and rated as normal sensitivity, hyposensitivity, or hypersensitivity. Pain and related characteristics were evaluated before TFESI, 1 week after and 4 weeks after intervention. RESULTS Seven sensory phenotypes were distinguished according to individual sets of sensory disturbances acquired with five sensory tests. Patients were grouped into four groups of allied phenotypes (normal sensitivity, hyposensitivity to 1 to 3 modalities, hyposensitivity to 4 to 5 modalities, and hypersensitivity). The whole study group showed significant improvement in most parameters of outcome measurements. Statistical analysis revealed some significant differences between sensory groups in patient-reported treatment effect scales after 4 weeks. We also detected significant differences between sensory groups in repeated measures after 1 and 4 weeks in some pain intensity characteristics and patient-reported treatment effect scales. CONCLUSION QualST might be clinically applicable for detecting patient subgroups that could differ in the early treatment results of radicular pain by TFESI.
Collapse
Affiliation(s)
- Alfredas Vaitkus
- Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,Centre of Anaesthesiology, Intensive Therapy and Pain Management, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Jūratė Šipylaitė
- Clinic of Anaesthesiology and Intensive Care, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,Centre of Anaesthesiology, Intensive Therapy and Pain Management, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| |
Collapse
|
36
|
Peripheral Mechanisms of Neuropathic Pain-the Role of Neuronal and Non-Neuronal Interactions and Their Implications for Topical Treatment of Neuropathic Pain. Pharmaceuticals (Basel) 2021; 14:ph14020077. [PMID: 33498496 PMCID: PMC7909513 DOI: 10.3390/ph14020077] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/13/2021] [Accepted: 01/16/2021] [Indexed: 12/16/2022] Open
Abstract
Neuropathic pain in humans arises as a consequence of injury or disease of somatosensory nervous system at peripheral or central level. Peripheral neuropathic pain is more common than central neuropathic pain, and is supposed to result from peripheral mechanisms, following nerve injury. The animal models of neuropathic pain show extensive functional and structural changes occurring in neuronal and non-neuronal cells in response to peripheral nerve injury. These pathological changes following damage lead to peripheral sensitization development, and subsequently to central sensitization initiation with spinal and supraspinal mechanism involved. The aim of this narrative review paper is to discuss the mechanisms engaged in peripheral neuropathic pain generation and maintenance, with special focus on the role of glial, immune, and epithelial cells in peripheral nociception. Based on the preclinical and clinical studies, interactions between neuronal and non-neuronal cells have been described, pointing out at the molecular/cellular underlying mechanisms of neuropathic pain, which might be potentially targeted by topical treatments in clinical practice. The modulation of the complex neuro-immuno-cutaneous interactions in the periphery represents a strategy for the development of new topical analgesics and their utilization in clinical settings.
Collapse
|
37
|
Cook DC, Goldstein PA. Non-canonical Molecular Targets for Novel Analgesics: Intracellular Calcium and HCN Channels. Curr Neuropharmacol 2021; 19:1937-1951. [PMID: 33463473 PMCID: PMC9185781 DOI: 10.2174/1570159x19666210119153047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/04/2021] [Accepted: 01/17/2021] [Indexed: 11/22/2022] Open
Abstract
Pain is a prevalent biopsychosocial condition that poses a significant challenge to healthcare providers, contributes substantially to a disability, and is a major economic burden worldwide. An overreliance on opioid analgesics, which primarily target the μ-opioid receptor, has caused devastating morbidity and mortality in the form of misuse and overdose-related death. Thus, novel analgesic medications are needed that can effectively treat pain and provide an alternative to opioids. A variety of cellular ion channels contribute to nociception, the response of the sensory nervous system to a noxious stimulus that commonly leads to pain. Ion channels involved in nociception may provide a suitable target for pharmacologic modulation to achieve pain relief. This narrative review summarizes the evidence for two ion channels that merit consideration as targets for non-opioid pain medications: ryanodine receptors (RyRs), which are intracellular calcium channels, and hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, which belong to the superfamily of voltage-gated K+ channels. The role of these channels in nociception and neuropathic pain is discussed and suitability as targets for novel analgesics and antihyperalgesics is considered.
Collapse
Affiliation(s)
- Daniel C. Cook
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Peter A. Goldstein
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY 10065, USA
- Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
38
|
An introduction to the Biennial Review of Pain. Pain 2020; 161 Suppl 1:S1-S2. [DOI: 10.1097/j.pain.0000000000001979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|