101
|
TRESK K + Channel Activity Regulates Trigeminal Nociception and Headache. eNeuro 2019; 6:ENEURO.0236-19.2019. [PMID: 31308053 PMCID: PMC6664143 DOI: 10.1523/eneuro.0236-19.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 06/23/2019] [Indexed: 02/07/2023] Open
Abstract
Although TWIK-related spinal cord K+ (TRESK) channel is expressed in all primary afferent neurons in trigeminal ganglia (TG) and dorsal root ganglia (DRG), whether TRESK activity regulates trigeminal pain processing is still not established. Dominant-negative TRESK mutations are associated with migraine but not with other types of pain in humans, suggesting that genetic TRESK dysfunction preferentially affects the generation of trigeminal pain, especially headache. Using TRESK global knock-out mice as a model system, we found that loss of TRESK in all TG neurons selectively increased the intrinsic excitability of small-diameter nociceptors, especially those that do not bind to isolectin B4 (IB4-). Similarly, loss of TRESK resulted in hyper-excitation of the small IB4- dural afferent neurons but not those that bind to IB4 (IB4+). Compared with wild-type littermates, both male and female TRESK knock-out mice exhibited more robust trigeminal nociceptive behaviors, including headache-related behaviors, whereas their body and visceral pain responses were normal. Interestingly, neither the total persistent outward current nor the intrinsic excitability was altered in adult TRESK knock-out DRG neurons, which may explain why genetic TRESK dysfunction is not associated with body and/or visceral pain in humans. We reveal for the first time that, among all primary afferent neurons, TG nociceptors are the most vulnerable to the genetic loss of TRESK. Our findings indicate that endogenous TRESK activity regulates trigeminal nociception, likely through controlling the intrinsic excitability of TG nociceptors. Importantly, we provide evidence that genetic loss of TRESK significantly increases the likelihood of developing headache.
Collapse
|
102
|
Andersen Hammond E, Pitz M, Shay B. Neuropathic Pain in Taxane-Induced Peripheral Neuropathy: Evidence for Exercise in Treatment. Neurorehabil Neural Repair 2019; 33:792-799. [PMID: 31342880 DOI: 10.1177/1545968319860486] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
One in 2 Canadians is expected to acquire cancer in their lifetime. Many cancers, including breast, ovarian, and lung cancer, are treated using taxane chemotherapy with curative intent. A major adverse effect with the use of taxane chemotherapeutic agents is taxane-induced peripheral neuropathy (TIPN). Both positive (spontaneous pain, heightened sensitivity with light touch, tingling, itching, burning) and negative (loss of touch, loss of hot/cold sensations, and loss of pain) sensory symptoms can be experienced in the hands and feet and worsen with increasing dose and treatment duration. The pathophysiology of TIPN is still unknown but likely involves multiple mechanisms, including microtubule impairment, neuroimmune and inflammatory changes, ion channel remodeling, impaired mitochondrial function, and genetic predisposition. This review highlights current theories on the pathophysiology for TIPN, the cellular responses thought to maintain neuropathic pain, and the growing support for exercise in the treatment and prevention of peripheral neuropathy and neuropathic pain in both animal and human models.
Collapse
|
103
|
Grotle AK, Crawford CK, Huo Y, Ybarbo KM, Harrison ML, Graham J, Stanhope KL, Havel PJ, Fadel PJ, Stone AJ. Exaggerated cardiovascular responses to muscle contraction and tendon stretch in UCD type-2 diabetes mellitus rats. Am J Physiol Heart Circ Physiol 2019; 317:H479-H486. [PMID: 31274351 DOI: 10.1152/ajpheart.00229.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Patients with type-2 diabetes mellitus (T2DM) have exaggerated sympathetic activity and blood pressure responses to exercise. However, the underlying mechanisms for these responses, as well as how these responses change throughout disease progression, are not completely understood. For this study, we examined the effect of the progression of T2DM on the exercise pressor reflex, a critical neurocardiovascular mechanism that functions to increase sympathetic activity and blood pressure during exercise. We also aimed to examine the effect of T2DM on reflexive cardiovascular responses to static contraction, as well as those responses to tendon stretch when an exaggerated exercise pressor reflex was present. We evoked the exercise pressor reflex and mechanoreflex by statically contracting the hindlimb muscles and stretching the Achilles tendon, respectively, for 30 s. We then compared pressor and cardioaccelerator responses in unanesthetized, decerebrated University of California Davis (UCD)-T2DM rats at 21 and 31 wk following the onset of T2DM to responses in healthy nondiabetic rats. We found that the pressor response to static contraction was greater in the 31-wk T2DM [change in mean arterial pressure (∆MAP) = 39 ± 5 mmHg] but not in the 21-wk T2DM (∆MAP = 24 ± 5 mmHg) rats compared with nondiabetic rats (∆MAP = 18 ± 2 mmHg; P < 0.05). Similarly, the pressor and the cardioaccelerator responses to tendon stretch were significantly greater in the 31-wk T2DM rats [∆MAP = 69 ± 6 mmHg; change in heart rate (∆HR) = 28 ± 4 beats/min] compared with nondiabetic rats (∆MAP = 14 ± 2 mmHg; ∆HR = 5 ± 3 beats/min; P < 0.05). These findings suggest that the exercise pressor reflex changes as T2DM progresses and that a sensitized mechanoreflex may play a role in exaggerating these cardiovascular responses.NEW & NOTEWORTHY This is the first study to provide evidence that as type-2 diabetes mellitus (T2DM) progresses, the exercise pressor reflex becomes exaggerated, an effect that may be due to a sensitized mechanoreflex. Moreover, these findings provide compelling evidence suggesting that impairments in the reflexive control of circulation contribute to exaggerated blood pressure responses to exercise in T2DM.
Collapse
Affiliation(s)
- Ann-Katrin Grotle
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Charles K Crawford
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Yu Huo
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Kai M Ybarbo
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Michelle L Harrison
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - James Graham
- Department of Molecular Biosciences School of Veterinary Medicine and Department of Nutrition; University of California Davis, Davis, California
| | - Kimber L Stanhope
- Department of Molecular Biosciences School of Veterinary Medicine and Department of Nutrition; University of California Davis, Davis, California
| | - Peter J Havel
- Department of Molecular Biosciences School of Veterinary Medicine and Department of Nutrition; University of California Davis, Davis, California
| | - Paul J Fadel
- Department of Kinesiology; The University of Texas at Arlington, Arlington, Texas
| | - Audrey J Stone
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
104
|
Abstract
The global epidemic of prediabetes and diabetes has led to a corresponding epidemic of complications of these disorders. The most prevalent complication is neuropathy, of which distal symmetric polyneuropathy (for the purpose of this Primer, referred to as diabetic neuropathy) is very common. Diabetic neuropathy is a loss of sensory function beginning distally in the lower extremities that is also characterized by pain and substantial morbidity. Over time, at least 50% of individuals with diabetes develop diabetic neuropathy. Glucose control effectively halts the progression of diabetic neuropathy in patients with type 1 diabetes mellitus, but the effects are more modest in those with type 2 diabetes mellitus. These findings have led to new efforts to understand the aetiology of diabetic neuropathy, along with new 2017 recommendations on approaches to prevent and treat this disorder that are specific for each type of diabetes. In parallel, new guidelines for the treatment of painful diabetic neuropathy using distinct classes of drugs, with an emphasis on avoiding opioid use, have been issued. Although our understanding of the complexities of diabetic neuropathy has substantially evolved over the past decade, the distinct mechanisms underlying neuropathy in type 1 and type 2 diabetes remains unknown. Future discoveries on disease pathogenesis will be crucial to successfully address all aspects of diabetic neuropathy, from prevention to treatment.
Collapse
|
105
|
Feldman EL, Callaghan BC, Pop-Busui R, Zochodne DW, Wright DE, Bennett DL, Bril V, Russell JW, Viswanathan V. Diabetic neuropathy. Nat Rev Dis Primers 2019; 5:42. [PMID: 31197183 PMCID: PMC7096070 DOI: 10.1038/s41572-019-0097-9] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The global epidemic of prediabetes and diabetes has led to a corresponding epidemic of complications of these disorders. The most prevalent complication is neuropathy, of which distal symmetric polyneuropathy (for the purpose of this Primer, referred to as diabetic neuropathy) is very common. Diabetic neuropathy is a loss of sensory function beginning distally in the lower extremities that is also characterized by pain and substantial morbidity. Over time, at least 50% of individuals with diabetes develop diabetic neuropathy. Glucose control effectively halts the progression of diabetic neuropathy in patients with type 1 diabetes mellitus, but the effects are more modest in those with type 2 diabetes mellitus. These findings have led to new efforts to understand the aetiology of diabetic neuropathy, along with new 2017 recommendations on approaches to prevent and treat this disorder that are specific for each type of diabetes. In parallel, new guidelines for the treatment of painful diabetic neuropathy using distinct classes of drugs, with an emphasis on avoiding opioid use, have been issued. Although our understanding of the complexities of diabetic neuropathy has substantially evolved over the past decade, the distinct mechanisms underlying neuropathy in type 1 and type 2 diabetes remains unknown. Future discoveries on disease pathogenesis will be crucial to successfully address all aspects of diabetic neuropathy, from prevention to treatment.
Collapse
Affiliation(s)
- Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.,
| | | | - Rodica Pop-Busui
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes (MEND), University of Michigan, Ann Arbor, MI, USA
| | - Douglas W. Zochodne
- Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Douglas E. Wright
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - David L. Bennett
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Vera Bril
- Division of Neurology, Department of Medicine, University of Toronto and University Health Network, Toronto, Ontario, Canada.,Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - James W. Russell
- Department of Neurology, University of Maryland and VA Maryland Health Care System, Baltimore, MD, USA
| | | |
Collapse
|
106
|
Yoshida A, Kurimoto S, Iwatsuki K, Saeki M, Nishizuka T, Nakano T, Yoneda H, Onishi T, Yamamoto M, Tatebe M, Hirata H. Upper extremity disability is associated with pain intensity and grip strength in women with bilateral idiopathic carpal tunnel syndrome. NeuroRehabilitation 2019; 44:199-205. [PMID: 30856127 DOI: 10.3233/nre-182589] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND The upper extremity disability in patients with carpal tunnel syndrome (CTS) is related to dysfunction due to the median nerve damage. However, there is no report on which dysfunctions affect the upper extremity disability. PURPOSE This study aimed to investigate which clinical factors influence upper extremity disability in women with CTS. METHODS We analyzed 60 hands of women with bilateral idiopathic CTS. Upper extremity disability was assessed using Hand10, a validated and self-administered tool. Pain intensity was measured using the Japanese version of the Short-Form McGill Pain Questionnaire (SF-MPQ-J). We performed nerve conduction studies, assessed physical and psychological parameters, and collected demographic data. Physical parameters comprised grip strength, pinch strength, tactile threshold, static 2-point discrimination sensation, and severity of numbness. Psychological parameters include depression, pain anxiety, and distress. RESULTS The bivariate analysis revealed that Hand10 was significantly correlated with age, symptom duration, SF-MPQ-J, grip strength, pain anxiety, and distress. Multiple regression analysis demonstrated that SF-MPQ-J and grip strength were related to Hand10 score. CONCLUSIONS Pain intensity and grip strength were dysfunctions affecting the upper extremity disability in women with bilateral idiopathic CTS. Rehabilitation approaches for CTS should be considered based on the adaptive activities of the neural networks.
Collapse
Affiliation(s)
- Akihito Yoshida
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Department of Rehabilitation, Nagoya University Hospital, Nagoya, Japan
| | - Shigeru Kurimoto
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Katsuyuki Iwatsuki
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Masaomi Saeki
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Takanobu Nishizuka
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Tomonori Nakano
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hidemasa Yoneda
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Tetsurou Onishi
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Michiro Yamamoto
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Masahiro Tatebe
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hitoshi Hirata
- Department of Hand Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
107
|
Price TJ, Ray PR. Recent advances toward understanding the mysteries of the acute to chronic pain transition. CURRENT OPINION IN PHYSIOLOGY 2019; 11:42-50. [PMID: 32322780 DOI: 10.1016/j.cophys.2019.05.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chronic pain affects up to a third of the population. Ongoing epidemiology studies suggest that the impact of chronic pain on the population is accelerating [1]. While advances have been made in understanding how chronic pain develops, there are still many important mysteries about how acute pain transitions to a chronic state. In this review, I summarize recent developments in the field with a focus on several areas of emerging research that are likely to have an important impact on the field. These include mechanisms of cellular plasticity that drive chronic pain, evidence of pervasive sex differential mechanisms in chronic pain and the profound impact that next generation sequencing technologies are having on this area of research.
Collapse
Affiliation(s)
- Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Pradipta R Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| |
Collapse
|
108
|
Basbaum A. Chemogenetic management of neuropathic pain. Brain 2019; 140:2522-2525. [PMID: 28969393 DOI: 10.1093/brain/awx227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Allan Basbaum
- Department of Anatomy, University California San Francisco, San Francisco, CA 94901, USA
| |
Collapse
|
109
|
Abstract
Phantom limb pain is a chronic neuropathic pain that develops in 45-85% of patients who undergo major amputations of the upper and lower extremities and appears predominantly during two time frames following an amputation: the first month and later about 1 year. Although in most patients the frequency and intensity of pain diminish over time, severe pain persists in about 5-10%. It has been proposed that factors in both the peripheral and central nervous systems play major roles in triggering the development and maintenance of pain associated with extremity amputations. Chronic pain is physically and mentally debilitating, affecting an individual's capacity for self-care, but also diminishing an individual's daily capacity for personal and economic independence. In addition, the pain may lead to depression and feelings of hopelessness. A National Center for Biotechnology Information study found that in the USA alone, the annual cost of dealing with neuropathic pain is more than $600 billion, with an estimated 20 million people in the USA suffering from this condition. Although the pain can be reduced by antiepileptic drugs and analgesics, they are frequently ineffective or their side effects preclude their use. The optimal approach for eliminating neuropathic pain and improving individuals' quality of life is the development of novel techniques that permanently prevent the development and maintenance of neuropathic pain, or that eliminate the pain once it has developed. What is still required is understanding when and where an effective novel technique must be applied, such as onto the nerve stump of the transected peripheral axons, dorsal root ganglion neurons, spinal cord, or cortex to induce the desired influences. This review, the second of two in this journal volume, examines the techniques that may be capable of reducing or eliminating chronic neuropathic pain once it has developed. Such an understanding will improve amputees' quality of life by blocking the mechanisms that trigger and/or maintain PLP and chronic neuropathic pain.
Collapse
Affiliation(s)
- Damien P Kuffler
- Institute of Neurobiology, University of Puerto Rico, Medical Science Campus, 201 Blvd. del Valle, San Juan, PR, 00901, Puerto Rico.
| |
Collapse
|
110
|
Meeker TJ, Keaser ML, Khan SA, Gullapalli RP, Seminowicz DA, Greenspan JD. Non-invasive Motor Cortex Neuromodulation Reduces Secondary Hyperalgesia and Enhances Activation of the Descending Pain Modulatory Network. Front Neurosci 2019; 13:467. [PMID: 31139047 PMCID: PMC6519323 DOI: 10.3389/fnins.2019.00467] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/24/2019] [Indexed: 11/29/2022] Open
Abstract
Central sensitization is a driving mechanism in many chronic pain patients, and manifests as hyperalgesia and allodynia beyond any apparent injury. Recent studies have demonstrated analgesic effects of motor cortex (M1) stimulation in several chronic pain disorders, yet its neural mechanisms remain uncertain. We evaluated whether anodal M1 transcranial direct current stimulation (tDCS) would mitigate central sensitization as measured by indices of secondary hyperalgesia. We used a capsaicin-heat pain model to elicit secondary mechanical hyperalgesia in 27 healthy subjects. In an assessor and subject-blind randomized, sham-controlled, crossover trial, anodal M1 tDCS decreased the intensity of pinprick hyperalgesia more than cathodal or sham tDCS. To elucidate the mechanism driving analgesia, subjects underwent fMRI of painful mechanical stimuli prior to and following induction of the pain model, after receiving M1 tDCS. We hypothesized that anodal M1 tDCS would enhance engagement of a descending pain modulatory (DPM) network in response to mechanical stimuli. Anodal tDCS normalized the effects of central sensitization on neurophysiological responses to mechanical pain in the medial prefrontal cortex, pregenual anterior cingulate cortex, and periaqueductal gray, important regions in the DPM network. Taken together, these results provide support for the hypothesis that anodal M1-tDCS reduces central sensitization-induced hyperalgesia through the DPM network in humans.
Collapse
Affiliation(s)
- Timothy J. Meeker
- Department of Neurosurgery, Johns Hopkins Medicine, Baltimore, MD, United States
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Baltimore, MD, United States
- Program in Neuroscience, University of Maryland, Baltimore, Baltimore, MD, United States
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Michael L. Keaser
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Shariq A. Khan
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Rao P. Gullapalli
- Program in Neuroscience, University of Maryland, Baltimore, Baltimore, MD, United States
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - David A. Seminowicz
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Baltimore, MD, United States
- Program in Neuroscience, University of Maryland, Baltimore, Baltimore, MD, United States
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Joel D. Greenspan
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Baltimore, MD, United States
- Program in Neuroscience, University of Maryland, Baltimore, Baltimore, MD, United States
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Baltimore, MD, United States
| |
Collapse
|
111
|
Grotle AK, Garcia EA, Harrison ML, Huo Y, Crawford CK, Ybarbo KM, Stone AJ. Exaggerated mechanoreflex in early-stage type 1 diabetic rats: role of Piezo channels. Am J Physiol Regul Integr Comp Physiol 2019; 316:R417-R426. [DOI: 10.1152/ajpregu.00294.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent findings have shown that muscle contraction evokes an exaggerated pressor response in type 1 diabetes mellitus (T1DM) rats; however, it is not known whether the mechanoreflex, which is commonly stimulated by stretching the Achilles tendon, contributes to this abnormal response. Furthermore, the role of mechano-gated Piezo channels, found on thin-fiber afferent endings, in evoking the mechanoreflex in T1DM is also unknown. Therefore, in male and female streptozotocin (STZ, 50 mg/kg)-induced T1DM and healthy control (CTL) rats, we examined the pressor and cardioaccelerator responses to tendon stretch during the early stage of the disease. To determine the role of Piezo channels, GsMTx-4, a selective Piezo channel inhibitor, was injected into the arterial supply of the hindlimb. At 1 wk after STZ injection in unanesthetized, decerebrate rats, we stretched the Achilles tendon for 30 s and measured pressor and cardioaccelerator responses. We then compared pressor and cardioaccelerator responses to tendon stretch before and after GsMTx-4 injection (10 µg/100 ml). We found that the pressor (change in mean arterial pressure) response [41 ± 5 mmHg ( n = 15) for STZ and 18 ± 3 mmHg ( n = 11) for CTL ( P < 0.01)] and cardioaccelerator (change in heart rate) response [18 ± 4 beats/min for STZ ( n = 15) and 8 ± 2 beats/min ( n = 11) for CTL ( P < 0.05)] to tendon stretch were exaggerated in STZ rats. Local injection of GsMTx-4 attenuated the pressor [55 ± 7 mmHg ( n = 6) before and 27 ± 9 mmHg ( n = 6) after GsMTx-4 ( P < 0.01)], but not the cardioaccelerator, response to tendon stretch in STZ rats and had no effect on either response in CTL rats. These data suggest that T1DM exaggerates the mechanoreflex response to tendon stretch and that Piezo channels play a role in this exaggeration.
Collapse
Affiliation(s)
- Ann-Katrin Grotle
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Elizabeth A. Garcia
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Michelle L. Harrison
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Yu Huo
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Charles K. Crawford
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Kai M. Ybarbo
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| | - Audrey J. Stone
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
112
|
Suppression of neuropathic pain by selective silencing of dorsal root ganglion ectopia using nonblocking concentrations of lidocaine. Pain 2019; 160:2105-2114. [DOI: 10.1097/j.pain.0000000000001602] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
113
|
Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-Hajj SD. The Role of Voltage-Gated Sodium Channels in Pain Signaling. Physiol Rev 2019; 99:1079-1151. [DOI: 10.1152/physrev.00052.2017] [Citation(s) in RCA: 462] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acute pain signaling has a key protective role and is highly evolutionarily conserved. Chronic pain, however, is maladaptive, occurring as a consequence of injury and disease, and is associated with sensitization of the somatosensory nervous system. Primary sensory neurons are involved in both of these processes, and the recent advances in understanding sensory transduction and human genetics are the focus of this review. Voltage-gated sodium channels (VGSCs) are important determinants of sensory neuron excitability: they are essential for the initial transduction of sensory stimuli, the electrogenesis of the action potential, and neurotransmitter release from sensory neuron terminals. Nav1.1, Nav1.6, Nav1.7, Nav1.8, and Nav1.9 are all expressed by adult sensory neurons. The biophysical characteristics of these channels, as well as their unique expression patterns within subtypes of sensory neurons, define their functional role in pain signaling. Changes in the expression of VGSCs, as well as posttranslational modifications, contribute to the sensitization of sensory neurons in chronic pain states. Furthermore, gene variants in Nav1.7, Nav1.8, and Nav1.9 have now been linked to human Mendelian pain disorders and more recently to common pain disorders such as small-fiber neuropathy. Chronic pain affects one in five of the general population. Given the poor efficacy of current analgesics, the selective expression of particular VGSCs in sensory neurons makes these attractive targets for drug discovery. The increasing availability of gene sequencing, combined with structural modeling and electrophysiological analysis of gene variants, also provides the opportunity to better target existing therapies in a personalized manner.
Collapse
Affiliation(s)
- David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Alex J. Clark
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Jianying Huang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G. Waxman
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Sulayman D. Dib-Hajj
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
114
|
Siouti E, Andreakos E. The many facets of macrophages in rheumatoid arthritis. Biochem Pharmacol 2019; 165:152-169. [PMID: 30910693 DOI: 10.1016/j.bcp.2019.03.029] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 03/19/2019] [Indexed: 12/24/2022]
Abstract
Macrophages are central to the pathophysiology of rheumatoid arthritis (RA). They constitute the main source of pro-inflammatory cytokines and chemokines such as TNF and IL-1β, they activate a wide range of immune and non-immune cells, and they secrete diverse tissue degrading enzymes driving chronic pro-inflammatory, tissue destructive and pain responses in RA. However, they can also produce anti-inflammatory cytokines such as IL-10, secrete inhibitors of tissue degrading enzymes and promote immunoregulatory and protective responses, suggesting the existence of macrophages with distinct and diverse functional activities. Although the underlying basis of this phenomenon has remained obscure for years, emerging evidence has now provided insight into the mechanisms and molecular processes involved. Here, we review current knowledge on the biology of macrophages in RA, and highlight recent literature on the heterogeneity, origins and ontogeny of macrophages as part of the mononuclear phagocyte system. We also discuss their plasticity in the context of the M1/M2 paradigm, and the emerging theme of metabolic rewiring as a major mechanism for programming macrophage functions and pro-inflammatory activities. This sheds light into the many facets of macrophages in RA, their molecular regulation and their translational potential for developing novel protective and therapeutic strategies in the clinic.
Collapse
Affiliation(s)
- Eleni Siouti
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; Airway Disease Infection Section, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London W2 1NY, United Kingdom.
| |
Collapse
|
115
|
Megat S, Ray PR, Moy JK, Lou TF, Barragán-Iglesias P, Li Y, Pradhan G, Wanghzou A, Ahmad A, Burton MD, North RY, Dougherty PM, Khoutorsky A, Sonenberg N, Webster KR, Dussor G, Campbell ZT, Price TJ. Nociceptor Translational Profiling Reveals the Ragulator-Rag GTPase Complex as a Critical Generator of Neuropathic Pain. J Neurosci 2019; 39:393-411. [PMID: 30459229 PMCID: PMC6335757 DOI: 10.1523/jneurosci.2661-18.2018] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/05/2018] [Accepted: 11/14/2018] [Indexed: 12/11/2022] Open
Abstract
Nociceptors, sensory neurons in the DRG that detect damaging or potentially damaging stimuli, are key drivers of neuropathic pain. Injury to these neurons causes activation of translation regulation signaling, including the mechanistic target of rapamycin complex 1 (mTORC1) and mitogen-activated protein kinase interacting kinase (MNK) eukaryotic initiation factor (eIF) 4E pathways. This is a mechanism driving changes in excitability of nociceptors that is critical for the generation of chronic pain states; however, the mRNAs that are translated to lead to this plasticity have not been elucidated. To address this gap in knowledge, we used translating ribosome affinity purification in male and female mice to comprehensively characterize mRNA translation in Scn10a-positive nociceptors in chemotherapy-induced neuropathic pain (CIPN) caused by paclitaxel treatment. This unbiased method creates a new resource for the field, confirms many findings in the CIPN literature and also find extensive evidence for new target mechanisms that may cause CIPN. We provide evidence that an underlying mechanism of CIPN is sustained mTORC1 activation driven by MNK1-eIF4E signaling. RagA, a GTPase controlling mTORC1 activity, is identified as a novel target of MNK1-eIF4E signaling. This demonstrates a novel translation regulation signaling circuit wherein MNK1-eIF4E activity drives mTORC1 via control of RagA translation. CIPN and RagA translation are strongly attenuated by genetic ablation of eIF4E phosphorylation, MNK1 elimination or treatment with the MNK inhibitor eFT508. We identify a novel translational circuit for the genesis of neuropathic pain caused by chemotherapy with important implications for therapeutics.SIGNIFICANCE STATEMENT Neuropathic pain affects up to 10% of the population, but its underlying mechanisms are incompletely understood, leading to poor treatment outcomes. We used translating ribosome affinity purification technology to create a comprehensive translational profile of DRG nociceptors in naive mice and at the peak of neuropathic pain induced by paclitaxel treatment. We reveal new insight into how mechanistic target of rapamycin complex 1 is activated in neuropathic pain pointing to a key role of MNK1-eIF4E-mediated translation of a complex of mRNAs that control mechanistic target of rapamycin complex 1 signaling at the surface of the lysosome. We validate this finding using genetic and pharmacological techniques. Our work strongly suggests that MNK1-eIF4E signaling drives CIPN and that a drug in human clinical trials, eFT508, may be a new therapeutic for neuropathic pain.
Collapse
Affiliation(s)
- Salim Megat
- University of Texas at Dallas, School of Behavioral and Brain Sciences, 800 Campbell Rd, Richardson, Texas, 75080
- University of Texas at Dallas, Center for Advanced Pain Studies, 800 Campbell Rd, Richardson, Texas, 75080
| | - Pradipta R Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences, 800 Campbell Rd, Richardson, Texas, 75080
- University of Texas at Dallas, Center for Advanced Pain Studies, 800 Campbell Rd, Richardson, Texas, 75080
| | - Jamie K Moy
- University of Texas at Dallas, School of Behavioral and Brain Sciences, 800 Campbell Rd, Richardson, Texas, 75080
| | - Tzu-Fang Lou
- University of Texas at Dallas, Department of Biological Sciences, 800 Campbell Rd, Richardson, Texas, 75080
| | - Paulino Barragán-Iglesias
- University of Texas at Dallas, School of Behavioral and Brain Sciences, 800 Campbell Rd, Richardson, Texas, 75080
- University of Texas at Dallas, Center for Advanced Pain Studies, 800 Campbell Rd, Richardson, Texas, 75080
| | - Yan Li
- University of Texas M.D. Anderson Cancer Center, Department of Anesthesia and Pain Medicine, 1400 Holcombe Boulevard, Houston, TX 77030
| | - Grishma Pradhan
- University of Texas at Dallas, School of Behavioral and Brain Sciences, 800 Campbell Rd, Richardson, Texas, 75080
- University of Texas at Dallas, Center for Advanced Pain Studies, 800 Campbell Rd, Richardson, Texas, 75080
| | - Andi Wanghzou
- University of Texas at Dallas, School of Behavioral and Brain Sciences, 800 Campbell Rd, Richardson, Texas, 75080
- University of Texas at Dallas, Center for Advanced Pain Studies, 800 Campbell Rd, Richardson, Texas, 75080
| | - Ayesha Ahmad
- University of Texas at Dallas, School of Behavioral and Brain Sciences, 800 Campbell Rd, Richardson, Texas, 75080
- University of Texas at Dallas, Center for Advanced Pain Studies, 800 Campbell Rd, Richardson, Texas, 75080
| | - Michael D Burton
- University of Texas at Dallas, School of Behavioral and Brain Sciences, 800 Campbell Rd, Richardson, Texas, 75080
- University of Texas at Dallas, Center for Advanced Pain Studies, 800 Campbell Rd, Richardson, Texas, 75080
| | - Robert Y North
- University of Texas M.D. Anderson Cancer Center, Department of Anesthesia and Pain Medicine, 1400 Holcombe Boulevard, Houston, TX 77030
| | - Patrick M Dougherty
- University of Texas M.D. Anderson Cancer Center, Department of Anesthesia and Pain Medicine, 1400 Holcombe Boulevard, Houston, TX 77030
| | - Arkady Khoutorsky
- McGill University, Department of Anesthesia, 001 Boulevard Décarie C05.2000, Montréal, QC H4A 3J1, Canada
| | - Nahum Sonenberg
- McGill University, Goodman Cancer Research Center, Department of Biochemistry, 1160 Pine Ave W, Montreal, QC H3A 1A3, Canada, and
| | - Kevin R Webster
- eFFECTOR Therapeutics, 11180 Roselle St, San Diego, CA 92121
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences, 800 Campbell Rd, Richardson, Texas, 75080
- University of Texas at Dallas, Center for Advanced Pain Studies, 800 Campbell Rd, Richardson, Texas, 75080
| | - Zachary T Campbell
- University of Texas at Dallas, Center for Advanced Pain Studies, 800 Campbell Rd, Richardson, Texas, 75080,
- University of Texas at Dallas, Department of Biological Sciences, 800 Campbell Rd, Richardson, Texas, 75080
| | - Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences, 800 Campbell Rd, Richardson, Texas, 75080,
- University of Texas at Dallas, Center for Advanced Pain Studies, 800 Campbell Rd, Richardson, Texas, 75080
| |
Collapse
|
116
|
Patel R, Kucharczyk M, Montagut‐Bordas C, Lockwood S, Dickenson AH. Neuropathy following spinal nerve injury shares features with the irritable nociceptor phenotype: A back-translational study of oxcarbazepine. Eur J Pain 2019; 23:183-197. [PMID: 30091265 PMCID: PMC6396087 DOI: 10.1002/ejp.1300] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/24/2018] [Accepted: 07/31/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND The term 'irritable nociceptor' was coined to describe neuropathic patients characterized by evoked hypersensitivity and preservation of primary afferent fibres. Oxcarbazepine is largely ineffectual in an overall patient population, but has clear efficacy in a subgroup with the irritable nociceptor profile. We examine whether neuropathy in rats induced by spinal nerve injury shares overlapping pharmacological sensitivity with the irritable nociceptor phenotype using drugs that target sodium channels. METHODS In vivo electrophysiology was performed in anaesthetized spinal nerve ligated (SNL) and sham-operated rats to record from wide dynamic range (WDR) neurones in the ventral posterolateral thalamus (VPL) and dorsal horn. RESULTS In neuropathic rats, spontaneous activity in the VPL was substantially attenuated by spinal lidocaine, an effect that was absent in sham rats. The former measure was in part dependent on ongoing peripheral activity as intraplantar lidocaine also reduced aberrant spontaneous thalamic firing. Systemic oxcarbazepine had no effect on wind-up of dorsal horn neurones in sham and SNL rats. However, in SNL rats, oxcarbazepine markedly inhibited punctate mechanical-, dynamic brush- and cold-evoked neuronal responses in the VPL and dorsal horn, with minimal effects on heat-evoked responses. In addition, oxcarbazepine inhibited spontaneous activity in the VPL. Intraplantar injection of the active metabolite licarbazepine replicated the effects of systemic oxcarbazepine, supporting a peripheral locus of action. CONCLUSIONS We provide evidence that ongoing activity in primary afferent fibres drives spontaneous thalamic firing after spinal nerve injury and that oxcarbazepine through a peripheral mechanism exhibits modality-selective inhibitory effects on sensory neuronal processing. SIGNIFICANCE The inhibitory effects of lidocaine and oxcarbazepine in this rat model of neuropathy resemble the clinical observations in the irritable nociceptor patient subgroup and support a mechanism-based rationale for bench-to-bedside translation when screening novel drugs.
Collapse
Affiliation(s)
- Ryan Patel
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | - Mateusz Kucharczyk
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | | | - Stevie Lockwood
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | - Anthony H. Dickenson
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
117
|
Freeman R, Edwards R, Baron R, Bruehl S, Cruccu G, Dworkin RH, Haroutounian S. AAPT Diagnostic Criteria for Peripheral Neuropathic Pain: Focal and Segmental Disorders. THE JOURNAL OF PAIN 2018; 20:369-393. [PMID: 30527971 DOI: 10.1016/j.jpain.2018.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 09/27/2018] [Accepted: 10/02/2018] [Indexed: 12/19/2022]
Abstract
Peripheral neuropathic pain is among the most prevalent types of neuropathic pain. No comprehensive peripheral neuropathic pain classification system that incorporates contemporary clinical, diagnostic, biological, and psychological information exists. To address this need, this article covers the taxonomy for 4 focal or segmental peripheral neuropathic pain disorders, as part of the Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks (ACTTION) public-private partnership and the American Pain Society (APS) collaborative to develop a standardized, evidence-based taxonomy initiative: the ACTTION-APS Pain Taxonomy (AAPT). The disorders-postherpetic neuralgia, persistent posttraumatic neuropathic pain, complex regional pain disorder, and trigeminal neuralgia-were selected because of their clinical and clinical research relevance. The multidimensional features of the taxonomy are suitable for clinical trials and can also facilitate hypothesis-driven case-control and cohort epidemiologic studies. PERSPECTIVE: The AAPT peripheral neuropathic pain taxonomy subdivides the peripheral neuropathic pain disorders into those that are generalized and symmetric and those that are focal or segmental and asymmetric. In this article, we cover the focal and segmental disorders: postherpetic neuralgia, persistent posttraumatic neuropathic pain, complex regional pain disorder, and trigeminal neuralgia. The taxonomy is evidence-based and multidimensional, with the following dimensions: 1) core diagnostic criteria; 2) common features; 3) common medical and psychiatric comorbidities; 4) neurobiological, psychosocial, and functional consequences; and 5) putative neurobiological and psychosocial mechanisms, risk factors, and protective factors.
Collapse
Affiliation(s)
- Roy Freeman
- Center for Autonomic and Peripheral Nerve Disorders, Beth Israel Deaconess Medical Center, Boston, MA.
| | - Robert Edwards
- Department of Anesthesiology, Brigham & Women's Hospital, Harvard University School of Medicine, Boston, MA
| | - Ralf Baron
- University of Kiel, Division of Neurological Pain Research and Therapy, Department of Neurology, Kiel, Germany
| | - Stephen Bruehl
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN
| | - Giorgio Cruccu
- Department Human Neuroscience, Sapienza University, Rome, Italy
| | - Robert H Dworkin
- Department of Anesthesiology and Perioperative Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Simon Haroutounian
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
118
|
Resilience to Pain: A Peripheral Component Identified Using Induced Pluripotent Stem Cells and Dynamic Clamp. J Neurosci 2018; 39:382-392. [PMID: 30459225 DOI: 10.1523/jneurosci.2433-18.2018] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/29/2018] [Accepted: 11/07/2018] [Indexed: 11/21/2022] Open
Abstract
Pain is a complex process that involves both detection in the peripheral nervous system and perception in the CNS. Individual-to-individual differences in pain are well documented, but not well understood. Here we capitalized on inherited erythromelalgia (IEM), a well characterized human genetic model of chronic pain, and studied a unique family containing related IEM subjects with the same disease-causing NaV1.7 mutation, which is known to make dorsal root ganglion (DRG) neurons hyperexcitable, but different pain profiles (affected son with severe pain, affected mother with moderate pain, and an unaffected father). We show, first, that, at least in some cases, relative sensitivity to pain can be modeled in subject-specific induced pluripotent stem cell (iPSC)-derived sensory neurons in vitro; second, that, in some cases, mechanisms operating in peripheral sensory neurons contribute to interindividual differences in pain; and third, using whole exome sequencing (WES) and dynamic clamp, we show that it is possible to pinpoint a specific variant of another gene, KCNQ in this particular kindred, that modulates the excitability of iPSC-derived sensory neurons in this family. While different gene variants may modulate DRG neuron excitability and thereby contribute to interindividual differences in pain in other families, this study shows that subject-specific iPSCs can be used to model interindividual differences in pain. We further provide proof-of-principle that iPSCs, WES, and dynamic clamp can be used to investigate peripheral mechanisms and pinpoint specific gene variants that modulate pain signaling and contribute to interindividual differences in pain.SIGNIFICANCE STATEMENT Individual-to-individual differences in pain are well documented, but not well understood. In this study, we show, first, that, at least in some cases, relative sensitivity to pain can be modeled in subject-specific induced pluripotent stem cell-derived sensory neurons in vitro; second, that, in some cases, mechanisms operating in peripheral sensory neurons contribute to interindividual differences in pain; and third, using whole exome sequencing and dynamic clamp, we show that it is possible to pinpoint a specific gene variant that modulates pain signaling and contributes to interindividual differences in pain.
Collapse
|
119
|
Black BJ, Atmaramani R, Plagens S, Campbell ZT, Dussor G, Price TJ, Pancrazio JJ. Emerging neurotechnology for antinoceptive mechanisms and therapeutics discovery. Biosens Bioelectron 2018; 126:679-689. [PMID: 30544081 DOI: 10.1016/j.bios.2018.11.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/01/2018] [Accepted: 11/10/2018] [Indexed: 12/20/2022]
Abstract
The tolerance, abuse, and potential exacerbation associated with classical chronic pain medications such as opioids creates a need for alternative therapeutics. Phenotypic screening provides a complementary approach to traditional target-based drug discovery. Profiling cellular phenotypes enables quantification of physiologically relevant traits central to a disease pathology without prior identification of a specific drug target. For complex disorders such as chronic pain, which likely involves many molecular targets, this approach may identify novel treatments. Sensory neurons, termed nociceptors, are derived from dorsal root ganglia (DRG) and can undergo changes in membrane excitability during chronic pain. In this review, we describe phenotypic screening paradigms that make use of nociceptor electrophysiology. The purpose of this paper is to review the bioelectrical behavior of DRG neurons, signaling complexity in sensory neurons, various sensory neuron models, assays for bioelectrical behavior, and emerging efforts to leverage microfabrication and microfluidics for assay development. We discuss limitations and advantages of these various approaches and offer perspectives on opportunities for future development.
Collapse
Affiliation(s)
- Bryan J Black
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA.
| | - Rahul Atmaramani
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Sarah Plagens
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Zachary T Campbell
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Joseph J Pancrazio
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| |
Collapse
|
120
|
Odem MA, Bavencoffe AG, Cassidy RM, Lopez ER, Tian J, Dessauer CW, Walters ET. Isolated nociceptors reveal multiple specializations for generating irregular ongoing activity associated with ongoing pain. Pain 2018; 159:2347-2362. [PMID: 30015712 PMCID: PMC6193853 DOI: 10.1097/j.pain.0000000000001341] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Ongoing pain has been linked to ongoing activity (OA) in human C-fiber nociceptors, but rodent models of pain-related OA have concentrated on allodynia rather than ongoing pain, and on OA generated in non-nociceptive Aβ fibers rather than C-fiber nociceptors. Little is known about how ongoing pain or nociceptor OA is generated. To define neurophysiological alterations underlying nociceptor OA, we have used isolated dorsal root ganglion neurons that continue to generate OA after removal from animals displaying ongoing pain. We subclassify OA as either spontaneous activity generated solely by alterations intrinsic to the active neuron or as extrinsically driven OA. Both types of OA were implicated previously in nociceptors in vivo and after isolation following spinal cord injury, which produces chronic ongoing pain. Using novel automated algorithms to analyze irregular changes in membrane potential, we have found, in a distinctive, nonaccommodating type of probable nociceptor, induction by spinal cord injury of 3 alterations that promote OA: (1) prolonged depolarization of resting membrane potential, (2) a hyperpolarizing shift in the voltage threshold for action potential generation, and (3) an increase in the incidence of large depolarizing spontaneous fluctuations (DSFs). Can DSFs also be enhanced acutely to promote OA in neurons from uninjured animals? A low dose of serotonin failed to change resting membrane potential but lowered action potential threshold. When combined with artificial depolarization to model inflammation, serotonin also strongly potentiated DSFs and OA. These findings reveal nociceptor specializations for generating OA that may promote ongoing pain in chronic and acute conditions.
Collapse
Affiliation(s)
- Max A. Odem
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Alexis G. Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Ryan M. Cassidy
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Elia R. Lopez
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| |
Collapse
|
121
|
McGaraughty S, Chu KL, Xu J. Characterization and pharmacological modulation of noci-responsive deep dorsal horn neurons across diverse rat models of pathological pain. J Neurophysiol 2018; 120:1893-1905. [DOI: 10.1152/jn.00325.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This overview compares the activity of wide dynamic range (WDR) and nociceptive specific (NS) neurons located in the deep dorsal horn across different rat models of pathological pain and following modulation by diverse pharmacology. The data were collected by our group under the same experimental conditions over numerous studies to facilitate comparison. Spontaneous firing of WDR neurons was significantly elevated (>3.7 Hz) in models of neuropathic, inflammation, and osteoarthritic pain compared with naive animals (1.9 Hz) but was very low (<0.5 Hz) and remained unchanged in NS neurons. WDR responses to low-intensity mechanical stimulation were elevated in neuropathic and inflammation models. WDR responses to high-intensity stimuli were enhanced in inflammatory (heat) and osteoarthritis (mechanical) models. NS responses to high-intensity stimulation did not change relative to control in any model examined. Several therapeutic agents reduced both evoked and spontaneous firing of WDR neurons (e.g., TRPV1, TRPV3, Nav1.7, Nav1.8, P2X7, P2X3, H3), other targets affected neither evoked nor spontaneous firing of WDR neurons (e.g., H4, TRPM8, KCNQ2/3), and some only modulated evoked (e.g, ASIC1a, Cav3.2) whereas others decreased evoked but affected spontaneous activity only in specific models (e.g., TRPA1, CB2). Spontaneous firing of WDR neurons was not altered by any peripherally restricted compound or by direct administration of compounds to peripheral sites, although the same compounds decreased evoked activity. Compounds acting centrally were effective against this endpoint. The diversity of incoming/modulating inputs to the deep dorsal horn positions this group of neurons as an important intersection within the pain system to validate novel therapeutics. NEW & NOTEWORTHY Data from multiple individual experiments were combined to show firing properties of wide dynamic range and nociceptive specific spinal dorsal horn neurons across varied pathological pain models. This high-powered analysis describes the sensitization following different forms of injury. Effects of diverse pharmacology on these neurons is also summarized from published and unpublished data all recorded under the same conditions to facilitate comparison. This comprehensive overview describes the function and utility of these neurons.
Collapse
Affiliation(s)
| | | | - Jun Xu
- AbbVie Discovery, North Chicago, Illinois
| |
Collapse
|
122
|
Structural, functional, and symptom relations in painful distal symmetric polyneuropathies: a systematic review. Pain 2018; 160:286-297. [DOI: 10.1097/j.pain.0000000000001381] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
123
|
Barry AM, Sondermann JR, Sondermann JH, Gomez-Varela D, Schmidt M. Region-Resolved Quantitative Proteome Profiling Reveals Molecular Dynamics Associated With Chronic Pain in the PNS and Spinal Cord. Front Mol Neurosci 2018; 11:259. [PMID: 30154697 PMCID: PMC6103001 DOI: 10.3389/fnmol.2018.00259] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/10/2018] [Indexed: 12/27/2022] Open
Abstract
To obtain a thorough understanding of chronic pain, large-scale molecular mapping of the pain axis at the protein level is necessary, but has not yet been achieved. We applied quantitative proteome profiling to build a comprehensive protein compendium of three regions of the pain neuraxis in mice: the sciatic nerve (SN), the dorsal root ganglia (DRG), and the spinal cord (SC). Furthermore, extensive bioinformatics analysis enabled us to reveal unique protein subsets which are specifically enriched in the peripheral nervous system (PNS) and SC. The immense value of these datasets for the scientific community is highlighted by validation experiments, where we monitored protein network dynamics during neuropathic pain. Here, we resolved profound region-specific differences and distinct changes of PNS-enriched proteins under pathological conditions. Overall, we provide a unique and validated systems biology proteome resource (summarized in our online database painproteome.em.mpg.de), which facilitates mechanistic insights into somatosensory biology and chronic pain—a prerequisite for the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Allison M Barry
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - Julia R Sondermann
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - Jan-Hendrik Sondermann
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - David Gomez-Varela
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | - Manuela Schmidt
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| |
Collapse
|
124
|
North RY, Lazaro TT, Dougherty PM. Ectopic Spontaneous Afferent Activity and Neuropathic Pain. Neurosurgery 2018; 65:49-54. [PMID: 31076785 DOI: 10.1093/neuros/nyy119] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/04/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Robert Y North
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Tyler T Lazaro
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Patrick M Dougherty
- The Departments of Pain Medicine Research, The Division of Anesthesia, Critical Care and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
125
|
Wijayasinghe N, Ringsted TK, Bischoff JM, Kehlet H, Werner MU. The role of peripheral afferents in persistent inguinal postherniorrhaphy pain: a randomized, double-blind, placebo-controlled, crossover trial of ultrasound-guided tender point blockade. Br J Anaesth 2018; 116:829-37. [PMID: 27199314 DOI: 10.1093/bja/aew071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2016] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Severe, persistent inguinal postherniorrhaphy pain (PIPP) is a debilitating condition that develops in 2-5% of patients. PIPP may be neuropathic in nature, yet the lesion in the peripheral nervous system has not been located. Most PIPP-patients demonstrate a tender point (TP) in the medial aspect of the inguinal region that triggers pain upon minimal pressure. As TPs may play a role in the pathophysiology of PIPP, the aim of this trial was to investigate the analgesic effects of local anaesthetic TP-blockade. METHODS A randomized, double-blind, placebo-controlled, crossover trial was performed in 14 PIPP-patients and six healthy volunteers. All participated in two sessions, seven days apart, receiving 10 ml of 0.25% bupivacaine or normal saline via an ultrasound-guided fascial plane block at the TP. The TP-area was used for pain assessments (at rest, on movement, with 100 kPa pressure-algometry) and quantitative sensory testing (pressure pain thresholds, thermal detection/pain thresholds, supra-threshold heat perception), before and after the TP-blockade. RESULTS The median (95% CI) reduction in pain was 63% (44.1 to 73.6%) after bupivacaine compared with 36% (11.6 to 49.7%; P=0.003) after placebo. Significant increases in cool detection (P=0.01) and pressure pain thresholds (P=0.009) with decreases in supra-threshold heat pain perception (P=0.003) were seen after bupivacaine only. In four out of six volunteers, increased thermal and evoked-pain thresholds after bupivacaine compared with placebo, was demonstrated. CONCLUSIONS This trial demonstrates that peripheral afferent input from the TP-area is important for maintenance of spontaneous and evoked pain in PIPP. CLINICAL TRIAL REGISTRATION NCT02065219.
Collapse
Affiliation(s)
| | - T K Ringsted
- Multidisciplinary Pain Center, Rigshospitalet, Copenhagen University Hospitals, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - J M Bischoff
- Multidisciplinary Pain Center, Rigshospitalet, Copenhagen University Hospitals, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - H Kehlet
- Section for Surgical Pathophysiology
| | - M U Werner
- Multidisciplinary Pain Center, Rigshospitalet, Copenhagen University Hospitals, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| |
Collapse
|
126
|
Do Changes in Sensory Processing Precede Low Back Pain Development in Healthy Individuals? Clin J Pain 2018; 34:525-531. [DOI: 10.1097/ajp.0000000000000563] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
127
|
Bernal L, Roza C. Hyperpolarization-activated channels shape temporal patterns of ectopic spontaneous discharge in C-nociceptors after peripheral nerve injury. Eur J Pain 2018; 22:1377-1387. [PMID: 29635758 DOI: 10.1002/ejp.1226] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Neuropathic pain is thought to be mediated by aberrant impulses from sensitized primary afferents, and the temporal summation of the discharges might also influence nociceptive processing. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels (Ih current) generate rhythmic activity in neurons within the central nervous system and contribute to nociceptors excitability in neuropathic pain. METHODS We searched for single fibres with ectopic spontaneous discharges from an in vitro preparation in mice containing a neuroma formed in a peripheral branch of the saphenous nerve together with the undamaged branches. RESULTS Both damaged (axotomized) and undamaged fibres (putative intact) developed ectopic spontaneous activity with different temporal spike trains: Clock-like, Irregular or Bursts. The Ih current blocker, ZD7288, significantly suppressed ectopic spontaneous discharges in nociceptive fibres (3/5 Aδ- and 24/31 C-units and 1 nonclassified) by 64%. Additionally, ZD7288 changed the spike patterns of 5/7 Clock-like and 3/4 Burst units to Irregular. Exogenous cAMP produced a significant ~65% increase in the ectopic firing in 5 Irregular fibres, which was restored by ZD7288. In six additional fibres (three Clock-like and three Irregular), exogenous cAMP had no further effect, but co-application with ZD7288 decreased their discharge by half. These units showed significant higher levels of discharges than the cAMP-sensitive ones. CONCLUSIONS Our data suggest that HCN channels modulate ectopic spontaneous firing in C-nociceptors and shape their temporal patterns of discharge which will, ultimately, modify the nociceptive message received and processed by second-order neurons. SIGNIFICANCE We show an involvement of HCN channels in the modulation of ectopic spontaneous discharges from C-nociceptors. This finding exposes a mechanism of nociceptive transmission enhancement and highlights the clinical relevance of peripheral HCN blockade for spontaneous pain relief during neuropathy.
Collapse
Affiliation(s)
- L Bernal
- Department of Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - C Roza
- Department of Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| |
Collapse
|
128
|
Kiguchi N, Kobayashi D, Saika F, Matsuzaki S, Kishioka S. Inhibition of peripheral macrophages by nicotinic acetylcholine receptor agonists suppresses spinal microglial activation and neuropathic pain in mice with peripheral nerve injury. J Neuroinflammation 2018; 15:96. [PMID: 29587798 PMCID: PMC5872578 DOI: 10.1186/s12974-018-1133-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 03/15/2018] [Indexed: 12/24/2022] Open
Abstract
Background Neuro–immune interaction underlies chronic neuroinflammation and aberrant sensory processing resulting in neuropathic pain. Despite the pathological significance of both neuroinflammation-driven peripheral sensitization and spinal sensitization, the functional relationship between these two distinct events has not been understood. Methods In this study, we determined whether inhibition of inflammatory macrophages by administration of α4β2 nicotinic acetylcholine receptor (nAChR) agonists improves neuropathic pain and affects microglial activation in the spinal dorsal horn (SDH) in mice following partial sciatic nerve ligation (PSL). Expression levels of neuroinflammatory molecules were evaluated by RT-qPCR and immunohistochemistry, and PSL-induced mechanical allodynia was defined by the von Frey test. Results Flow cytometry revealed that CD11b+ F4/80+ macrophages were accumulated in the injured sciatic nerve (SCN) after PSL. TC-2559, a full agonist for α4β2 nAChR, suppressed the upregulation of interleukin-1β (IL-1β) in the injured SCN after PSL and attenuated lipopolysaccharide-induced upregulation of IL-1β in cultured macrophages. Systemic (subcutaneous, s.c.) administration of TC-2559 during either the early (days 0–3) or middle/late (days 7–10) phase of PSL improved mechanical allodynia. Moreover, local (perineural, p.n.) administration of TC-2559 and sazetidine A, a partial agonist for α4β2 nAChR, during either the early or middle phase of PSL improved mechanical allodynia. However, p.n. administration of sazetidine A during the late (days 21–24) phase did not show the attenuating effect, whereas p.n. administration of TC-2559 during this phase relieved mechanical allodynia. Most importantly, p.n. administration of TC-2559 significantly suppressed morphological activation of Iba1+ microglia and decreased the upregulation of inflammatory microglia-dominant molecules, such as CD68, interferon regulatory factor 5, and IL-1β in the SDH after PSL. Conclusion These findings support the notion that pharmacological inhibition of inflammatory macrophages using an α4β2 nAChR agonist exhibit a wide therapeutic window on neuropathic pain after nerve injury, and it could be nominated as a novel pharmacotherapy to relieve intractable pain. Electronic supplementary material The online version of this article (10.1186/s12974-018-1133-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Pharmacology, Wakayama Medical University, 811-1 Kimiidera, Wakayama city, Wakayama, 641-0012, Japan.
| | - Daichi Kobayashi
- Department of Pharmacology, Wakayama Medical University, 811-1 Kimiidera, Wakayama city, Wakayama, 641-0012, Japan
| | - Fumihiro Saika
- Department of Pharmacology, Wakayama Medical University, 811-1 Kimiidera, Wakayama city, Wakayama, 641-0012, Japan
| | - Shinsuke Matsuzaki
- Department of Pharmacology, Wakayama Medical University, 811-1 Kimiidera, Wakayama city, Wakayama, 641-0012, Japan
| | - Shiroh Kishioka
- Department of Pharmacology, Wakayama Medical University, 811-1 Kimiidera, Wakayama city, Wakayama, 641-0012, Japan
| |
Collapse
|
129
|
How central is central poststroke pain? The role of afferent input in poststroke neuropathic pain: a prospective, open-label pilot study. Pain 2018; 159:1317-1324. [DOI: 10.1097/j.pain.0000000000001213] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
130
|
Megat S, Price TJ. Therapeutic opportunities for pain medicines via targeting of specific translation signaling mechanisms. NEUROBIOLOGY OF PAIN 2018; 4:8-19. [PMID: 30211342 PMCID: PMC6130820 DOI: 10.1016/j.ynpai.2018.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A common underlying cause of chronic pain is a phenotypic change in nociceptors in the peripheral nervous system. Translation regulation signaling pathways control gene expression changes that drive chronic pain. We focus on developments in pharmacology around translation regulation signaling that may yield new pain therapeutics.
As the population of the world ages and as more and more people survive diseases that used to be primary causes of mortality, the incidence of severe chronic pain in most of the world has risen dramatically. This type of pain is very difficult to treat and the opioid overdose epidemic that has become a leading cause of death in the United States and other parts of the world highlights the urgent need to develop new pain therapeutics. A common underlying cause of severe chronic pain is a phenotypic change in pain-sensing neurons in the peripheral nervous system called nociceptors. These neurons play a vital role in detecting potentially injurious stimuli, but when these neurons start to detect very low levels of inflammatory meditators or become spontaneously active, they send spurious pain signals to the brain that are significant drivers of chronic pain. An important question is what drives this phenotypic shift in nociceptors from quiescence under most conditions to sensitization to a broad variety of stimuli and spontaneous activity. The goal of this review is to discuss the critical role that specific translation regulation signaling pathways play in controlling gene expression changes that drive nociceptor sensitization and may underlie the development of spontaneous activity. The focus will be on advances in technologies that allow for identification of such targets and on developments in pharmacology around translation regulation signaling that may yield new pain therapeutics. A key advantage of pharmacological manipulation of these signaling events is that they may reverse phenotypic shifts in nociceptors that drive chronic pain thereby creating the first generation of disease modifying drugs for chronic pain.
Collapse
Affiliation(s)
- Salim Megat
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, USA
| |
Collapse
|
131
|
Meacham K, Shepherd A, Mohapatra DP, Haroutounian S. Neuropathic Pain: Central vs. Peripheral Mechanisms. Curr Pain Headache Rep 2018; 21:28. [PMID: 28432601 DOI: 10.1007/s11916-017-0629-5] [Citation(s) in RCA: 284] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW Our goal is to examine the processes-both central and peripheral-that underlie the development of peripherally-induced neuropathic pain (pNP) and to highlight recent evidence for mechanisms contributing to its maintenance. While many pNP conditions are initiated by damage to the peripheral nervous system (PNS), their persistence appears to rely on maladaptive processes within the central nervous system (CNS). The potential existence of an autonomous pain-generating mechanism in the CNS creates significant implications for the development of new neuropathic pain treatments; thus, work towards its resolution is crucial. Here, we seek to identify evidence for PNS and CNS independently generating neuropathic pain signals. RECENT FINDINGS Recent preclinical studies in pNP support and provide key details concerning the role of multiple mechanisms leading to fiber hyperexcitability and sustained electrical discharge to the CNS. In studies regarding central mechanisms, new preclinical evidence includes the mapping of novel inhibitory circuitry and identification of the molecular basis of microglia-neuron crosstalk. Recent clinical evidence demonstrates the essential role of peripheral mechanisms, mostly via studies that block the initially damaged peripheral circuitry. Clinical central mechanism studies use imaging to identify potentially self-sustaining infra-slow CNS oscillatory activity that may be unique to pNP patients. While new preclinical evidence supports and expands upon the key role of central mechanisms in neuropathic pain, clinical evidence for an autonomous central mechanism remains relatively limited. Recent findings from both preclinical and clinical studies recapitulate the critical contribution of peripheral input to maintenance of neuropathic pain. Further clinical investigations on the possibility of standalone central contributions to pNP may be assisted by a reconsideration of the agreed terms or criteria for diagnosing the presence of central sensitization in humans.
Collapse
Affiliation(s)
- Kathleen Meacham
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew Shepherd
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Durga P Mohapatra
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Simon Haroutounian
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
132
|
Adi T, Estacion M, Schulman BR, Vernino S, Dib-Hajj SD, Waxman SG. A novel gain-of-function Na v1.7 mutation in a carbamazepine-responsive patient with adult-onset painful peripheral neuropathy. Mol Pain 2018; 14:1744806918815007. [PMID: 30392441 PMCID: PMC6856981 DOI: 10.1177/1744806918815007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/12/2018] [Indexed: 12/13/2022] Open
Abstract
Voltage-gated sodium channel Nav1.7 is a threshold channel in peripheral dorsal root ganglion (DRG), trigeminal ganglion, and sympathetic ganglion neurons. Gain-of-function mutations in Nav1.7 have been shown to increase excitability in DRG neurons and have been linked to rare Mendelian and more common pain disorders. Discovery of Nav1.7 variants in patients with pain disorders may expand the spectrum of painful peripheral neuropathies associated with a well-defined molecular target, thereby providing a basis for more targeted approaches for treatment. We screened the genome of a patient with adult-onset painful peripheral neuropathy characterized by severe burning pain and report here the new Nav1.7-V810M variant. Voltage-clamp recordings were used to assess the effects of the mutation on biophysical properties of Nav1.7 and the response of the mutant channel to treatment with carbamazepine (CBZ), and multi-electrode array (MEA) recordings were used to assess the effects of the mutation on the excitability of neonatal rat pup DRG neurons. The V810M variant increases current density, shifts activation in a hyperpolarizing direction, and slows kinetics of deactivation, all gain-of-function attributes. We also show that DRG neurons that express the V810M variant become hyperexcitable. The patient responded to treatment with CBZ. Although CBZ did not depolarize activation of the mutant channel, it enhanced use-dependent inhibition. Our results demonstrate the presence of a novel gain-of-function variant of Nav1.7 in a patient with adult-onset painful peripheral neuropathy and the responsiveness of that patient to treatment with CBZ, which is likely due to the classical mechanism of use-dependent inhibition.
Collapse
Affiliation(s)
- Talia Adi
- Department of Neurology, Yale University School of Medicine, New
Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans
Affairs Medical Center, West Haven, CT, USA
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New
Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans
Affairs Medical Center, West Haven, CT, USA
| | - Betsy R Schulman
- Department of Neurology, Yale University School of Medicine, New
Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans
Affairs Medical Center, West Haven, CT, USA
| | - Steven Vernino
- Department of Neurology and Neurotherapeutics, UT Southwestern
Medical Center, Dallas, TX, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New
Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans
Affairs Medical Center, West Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New
Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans
Affairs Medical Center, West Haven, CT, USA
| |
Collapse
|
133
|
Abstract
The sensation of pain plays a vital protecting role, alerting organisms about potentially damaging stimuli. Tissue injury is detected by nerve endings of specialized peripheral sensory neurons called nociceptors that are equipped with different ion channels activated by thermal, mechanic, and chemical stimuli. Several transient receptor potential channels have been identified as molecular transducers of thermal stimuli in pain-sensing neurons. Skin injury or inflammation leads to increased sensitivity to thermal and mechanic stimuli, clinically defined as allodynia or hyperalgesia. This hypersensitivity is also characteristic of systemic inflammatory disorders and neuropathic pain conditions. Mechanisms of thermal hyperalgesia include peripheral sensitization of nociceptor afferents and maladaptive changes in pain-encoding neurons within the central nervous system. An important aspect of pain management involves attempts to minimize the development of nociceptor hypersensitivity. However, knowledge about the cellular and molecular mechanisms causing thermal hyperalgesia and allodynia in human subjects is still limited, and such knowledge would be an essential step for the development of more effective therapies.
Collapse
Affiliation(s)
- Félix Viana
- Alicante Institute of Neurosciences, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Spain.
| |
Collapse
|
134
|
Cruccu G, Nurmikko TJ, Ernault E, Riaz FK, McBride WT, Haanpää M. Superiority of capsaicin 8% patch versus oral pregabalin on dynamic mechanical allodynia in patients with peripheral neuropathic pain. Eur J Pain 2017; 22:700-706. [PMID: 29194851 PMCID: PMC5887877 DOI: 10.1002/ejp.1155] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2017] [Indexed: 01/17/2023]
Abstract
Background Dynamic Mechanical Allodynia (DMA) is a typical symptom of neuropathic pain (NP). In a recent study, the capsaicin 8% patch was noninferior to pregabalin in overall peripheral NP relief. In this study, we report the comparison of the two treatments in relieving DMA. Methods In a randomized, open‐label, head‐to‐head, 8‐week study, 488 patients with peripheral NP were treated with the capsaicin 8% patch (one application) or an optimized dose of pregabalin. Assessments included the area and intensity of DMA, and the number of patients achieving complete resolution of DMA. Results At baseline, 253 patients in the capsaicin 8% patch group and 235 patients in the pregabalin group had DMA. From baseline to end of study, the change in DMA intensity was significantly in favour of the capsaicin 8% patch versus pregabalin [−0.63 (95% CI: −1.04, −0.23; p = 0.002)]. Similarly, the capsaicin 8% patch was superior to pregabalin in reducing the area of DMA [−39.5 cm2 (95% CI: −69.1, −10.0; p = 0.009)] from baseline to end of study. Overall, a greater proportion of patients had a complete resolution of allodynia with capsaicin 8% patch treatment compared with pregabalin treatment (24.1% vs. 12.3%; p = 0.001) at end of study. Conclusion Capsaicin 8% treatment was superior to pregabalin in reducing the intensity and area of DMA, and in the number of patients with complete resolution of DMA. Significance The superiority of a topical treatment over pregabalin in relieving DMA supports the view that both peripheral and central sensitization can mediate allodynia.
Collapse
Affiliation(s)
- G Cruccu
- Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - T J Nurmikko
- The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - E Ernault
- Astellas Pharma Inc., Leiden, The Netherlands
| | - F K Riaz
- Astellas Pharma Inc., Chertsey, UK
| | - W T McBride
- Belfast Health and Social Care Trust, Belfast, Northern Ireland
| | - M Haanpää
- Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
135
|
Abstract
Diabetic polyneuropathy (DPN) is a major cause of neuropathic pain and a frequent target condition in analgesic treatment trials. Differences in the clinical symptoms and signs associated with DPN suggest distinct pathophysiological mechanisms underlying nerve damage and dysfunction that are likely to have therapeutic relevance. The aim of this study was to develop a tool for the bedside assessment of painful neuropathies such as DPN that captures the diversity of phenotypes. Sixty-one patients with type 2 diabetes and painful neuropathy, 19 patients with painless DPN, 25 patients with type 2 diabetes but no clinical evidence of neuropathy, and 20 healthy control subjects completed a structured interview (47 items) and a standardized physical examination (39 items). After analyzing critical features of pain and painless symptoms and examining the outcome of physical tests of sensory function, we determined principal components of the phenotypic variance among patients. Increased sensitivity to mechanical or thermal stimuli and, to a lesser extent, the sensory quality of pain or paresthesia were the most discriminating elements of DPN phenotypes. Correlation patterns of symptoms and signs indicated the involvement of functionally distinct nerve fiber populations. We combined interview questions and physical tests identifying these differences in a shortened assessment protocol that we named Standardized Evaluation of Pain and Somatosensory Function (StEPS). The protocol StEPS generates a phenotypic profile of patients with neuropathy. Separate intensity ratings for spontaneous painful symptoms and pain evoked by standard stimuli support a detailed documentation of neuropathic pain and its response to analgesic treatment.
Collapse
|
136
|
Kiguchi N, Kobayashi D, Saika F, Matsuzaki S, Kishioka S. Pharmacological Regulation of Neuropathic Pain Driven by Inflammatory Macrophages. Int J Mol Sci 2017; 18:ijms18112296. [PMID: 29104252 PMCID: PMC5713266 DOI: 10.3390/ijms18112296] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 10/27/2017] [Accepted: 10/31/2017] [Indexed: 12/16/2022] Open
Abstract
Neuropathic pain can have a major effect on quality of life but current therapies are often inadequate. Growing evidence suggests that neuropathic pain induced by nerve damage is caused by chronic inflammation. Upon nerve injury, damaged cells secrete pro-inflammatory molecules that activate cells in the surrounding tissue and recruit circulating leukocytes to the site of injury. Among these, the most abundant cell type is macrophages, which produce several key molecules involved in pain enhancement, including cytokines and chemokines. Given their central role in the regulation of peripheral sensitization, macrophage-derived cytokines and chemokines could be useful targets for the development of novel therapeutics. Inhibition of key pro-inflammatory cytokines and chemokines prevents neuroinflammation and neuropathic pain; moreover, recent studies have demonstrated the effectiveness of pharmacological inhibition of inflammatory (M1) macrophages. Nicotinic acetylcholine receptor ligands and T helper type 2 cytokines that reduce M1 macrophages are able to relieve neuropathic pain. Future translational studies in non-human primates will be crucial for determining the regulatory mechanisms underlying neuroinflammation-associated neuropathic pain. In turn, this knowledge will assist in the development of novel pharmacotherapies targeting macrophage-driven neuroinflammation for the treatment of intractable neuropathic pain.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Pharmacology, Wakayama Medical University, Wakayama 641-0012, Japan.
| | - Daichi Kobayashi
- Department of Pharmacology, Wakayama Medical University, Wakayama 641-0012, Japan.
| | - Fumihiro Saika
- Department of Pharmacology, Wakayama Medical University, Wakayama 641-0012, Japan.
| | - Shinsuke Matsuzaki
- Department of Pharmacology, Wakayama Medical University, Wakayama 641-0012, Japan.
| | - Shiroh Kishioka
- Department of Pharmacology, Wakayama Medical University, Wakayama 641-0012, Japan.
| |
Collapse
|
137
|
Towards a neurobiological understanding of pain in chronic pancreatitis: mechanisms and implications for treatment. Pain Rep 2017; 2:e625. [PMID: 29392239 PMCID: PMC5741325 DOI: 10.1097/pr9.0000000000000625] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 08/30/2017] [Accepted: 09/08/2017] [Indexed: 12/19/2022] Open
Abstract
We summarize the evidence for a neurobiological understanding of pain in patients with chronic pancreatitis and discuss its potential impact on prevention and treatment. Introduction: Chronic pancreatitis (CP) is a disease characterized by inflammation of the pancreas resulting in replacement of the normal functioning parenchyma by fibrotic connective tissue. This process leads to progressively impairment of exocrine and endocrine function and many patients develop a chronic pain syndrome. Objectives: We aimed to characterize the neurobiological signature of pain associated with CP and to discuss its implications for treatment strategies. Methods: Relevant basic and clinical articles were selected for review following an extensive search of the literature. Results: Pathophysiological changes in the peripheral (pancreatic gland) and central nervous system characterize the pain syndrome associated with CP; involved mechanisms can be broken down to 3 main branches: (1) peripheral sensitization, (2) pancreatic neuropathy, and (3) neuroplastic changes in the central pain pathways. Disease flares (recurrent pancreatitis) may accelerate the pathophysiological process and further sensitize the pain system, which ultimately results in an autonomous and self-perpetuating pain state that may become independent of the peripheral nociceptive drive. These findings share many similarities with those observed in neuropathic pain disorders and have important implications for treatment; adjuvant analgesics are effective in a subset of patients, and neuromodulation and neuropsychological interventions may prove useful in the future. Conclusion: Chronic pancreatitis is associated with abnormal processing of pain at the peripheral and central level of the pain system. This neurobiological understanding of pain has important clinical implications for treatment and prevention of pain chronification.
Collapse
|
138
|
Weir GA, Middleton SJ, Clark AJ, Daniel T, Khovanov N, McMahon SB, Bennett DL. Using an engineered glutamate-gated chloride channel to silence sensory neurons and treat neuropathic pain at the source. Brain 2017; 140:2570-2585. [PMID: 28969375 PMCID: PMC5841150 DOI: 10.1093/brain/awx201] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/12/2017] [Accepted: 07/03/2017] [Indexed: 12/14/2022] Open
Abstract
See Basbaum (doi:10.1093/brain/awx227) for a scientific commentary on this article. Peripheral neuropathic pain arises as a consequence of injury to sensory neurons; the development of ectopic activity in these neurons is thought to be critical for the induction and maintenance of such pain. Local anaesthetics and anti-epileptic drugs can suppress hyperexcitability; however, these drugs are complicated by unwanted effects on motor, central nervous system and cardiac function, and alternative more selective treatments to suppress hyperexcitability are therefore required. Here we show that a glutamate-gated chloride channel modified to be activated by low doses of ivermectin (but not glutamate) is highly effective in silencing sensory neurons and reversing neuropathic pain-related hypersensitivity. Activation of the glutamate-gated chloride channel expressed in either rodent or human induced pluripotent stem cell-derived sensory neurons in vitro potently inhibited their response to both electrical and algogenic stimuli. We have shown that silencing is achieved both at nerve terminals and the soma and is independent of membrane hyperpolarization and instead likely mediated by lowering of the membrane resistance. Using intrathecal adeno-associated virus serotype 9-based delivery, the glutamate-gated chloride channel was successfully targeted to mouse sensory neurons in vivo, resulting in high level and long-lasting expression of the channel selectively in sensory neurons. This enabled reproducible and reversible modulation of thermal and mechanical pain thresholds in vivo; analgesia was observed for 3 days after a single systemic dose of ivermectin. We did not observe any motor or proprioceptive deficits and noted no reduction in cutaneous afferent innervation or upregulation of the injury marker ATF3 following prolonged glutamate-gated chloride channel expression. Established mechanical and cold pain-related hypersensitivity generated by the spared nerve injury model of neuropathic pain was reversed by ivermectin treatment. The efficacy of ivermectin in ameliorating behavioural hypersensitivity was mirrored at the cellular level by a cessation of ectopic activity in sensory neurons. These findings demonstrate the importance of aberrant afferent input in the maintenance of neuropathic pain and the potential for targeted chemogenetic silencing as a new treatment modality in neuropathic pain.
Collapse
Affiliation(s)
- Greg A Weir
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Alex J Clark
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Tarun Daniel
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | | | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
139
|
MicroRNA cluster miR-17-92 regulates multiple functionally related voltage-gated potassium channels in chronic neuropathic pain. Nat Commun 2017; 8:16079. [PMID: 28677679 PMCID: PMC5504285 DOI: 10.1038/ncomms16079] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 05/26/2017] [Indexed: 12/18/2022] Open
Abstract
miR-17-92 is a microRNA cluster with six distinct members. Here, we show that the miR-17-92 cluster and its individual members modulate chronic neuropathic pain. All cluster members are persistently upregulated in primary sensory neurons after nerve injury. Overexpression of miR-18a, miR-19a, miR-19b and miR-92a cluster members elicits mechanical allodynia in rats, while their blockade alleviates mechanical allodynia in a rat model of neuropathic pain. Plausible targets for the miR-17-92 cluster include genes encoding numerous voltage-gated potassium channels and their modulatory subunits. Single-cell analysis reveals extensive co-expression of miR-17-92 cluster and its predicted targets in primary sensory neurons. miR-17-92 downregulates the expression of potassium channels, and reduced outward potassium currents, in particular A-type currents. Combined application of potassium channel modulators synergistically alleviates mechanical allodynia induced by nerve injury or miR-17-92 overexpression. miR-17-92 cluster appears to cooperatively regulate the function of multiple voltage-gated potassium channel subunits, perpetuating mechanical allodynia. Dysregulation of voltage gated potassium channels is a feature of neuropathic pain. Here in a rat model the authors identify the microRNA cluster miR-17-92 as a regulator of voltage gated potassium channels in the dorsal root ganglion neurons.
Collapse
|
140
|
Kornecook TJ, Yin R, Altmann S, Be X, Berry V, Ilch CP, Jarosh M, Johnson D, Lee JH, Lehto SG, Ligutti J, Liu D, Luther J, Matson D, Ortuno D, Roberts J, Taborn K, Wang J, Weiss MM, Yu V, Zhu DXD, Fremeau RT, Moyer BD. Pharmacologic Characterization of AMG8379, a Potent and Selective Small Molecule Sulfonamide Antagonist of the Voltage-Gated Sodium Channel Na V1.7. J Pharmacol Exp Ther 2017; 362:146-160. [PMID: 28473457 DOI: 10.1124/jpet.116.239590] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/03/2017] [Indexed: 03/08/2025] Open
Abstract
Potent and selective antagonists of the voltage-gated sodium channel NaV1.7 represent a promising avenue for the development of new chronic pain therapies. We generated a small molecule atropisomer quinolone sulfonamide antagonist AMG8379 and a less active enantiomer AMG8380. Here we show that AMG8379 potently blocks human NaV1.7 channels with an IC50 of 8.5 nM and endogenous tetrodotoxin (TTX)-sensitive sodium channels in dorsal root ganglion (DRG) neurons with an IC50 of 3.1 nM in whole-cell patch clamp electrophysiology assays using a voltage protocol that interrogates channels in a partially inactivated state. AMG8379 was 100- to 1000-fold selective over other NaV family members, including NaV1.4 expressed in muscle and NaV1.5 expressed in the heart, as well as TTX-resistant NaV channels in DRG neurons. Using an ex vivo mouse skin-nerve preparation, AMG8379 blocked mechanically induced action potential firing in C-fibers in both a time-dependent and dose-dependent manner. AMG8379 similarly reduced the frequency of thermally induced C-fiber spiking, whereas AMG8380 affected neither mechanical nor thermal responses. In vivo target engagement of AMG8379 in mice was evaluated in multiple NaV1.7-dependent behavioral endpoints. AMG8379 dose-dependently inhibited intradermal histamine-induced scratching and intraplantar capsaicin-induced licking, and reversed UVB radiation skin burn-induced thermal hyperalgesia; notably, behavioral effects were not observed with AMG8380 at similar plasma exposure levels. AMG8379 is a potent and selective NaV1.7 inhibitor that blocks sodium current in heterologous cells as well as DRG neurons, inhibits action potential firing in peripheral nerve fibers, and exhibits pharmacodynamic effects in translatable models of both itch and pain.
Collapse
Affiliation(s)
- Thomas J Kornecook
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Ruoyuan Yin
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Stephen Altmann
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Xuhai Be
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Virginia Berry
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Christopher P Ilch
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Michael Jarosh
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Danielle Johnson
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Josie H Lee
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Sonya G Lehto
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Joseph Ligutti
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Dong Liu
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Jason Luther
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - David Matson
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Danny Ortuno
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - John Roberts
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Kristin Taborn
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Jinti Wang
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Matthew M Weiss
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Violeta Yu
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Dawn X D Zhu
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Robert T Fremeau
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| | - Bryan D Moyer
- Department of Neuroscience (T.J.K., R.Y., S.A, C.P.I., M.J., D.J., J.H.L., S.G.L., J.Li., D.L., J.Lu., D.M., D.O., K.T., J.W., V.Y., D.X.D.Z., R.T.F., B.D.M.), Department of Medicinal Chemistry (M.M.W.), and Department of Pharmacokinetics and Drug Metabolism (X.B., V.B., J.R.), Amgen Inc., Cambridge, Massachusetts and Thousand Oaks, California
| |
Collapse
|
141
|
Andersen KG, Duriaud HM, Kehlet H, Aasvang EK. The Relationship Between Sensory Loss and Persistent Pain 1 Year After Breast Cancer Surgery. THE JOURNAL OF PAIN 2017; 18:1129-1138. [PMID: 28502878 DOI: 10.1016/j.jpain.2017.05.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 03/07/2017] [Accepted: 05/04/2017] [Indexed: 10/19/2022]
Abstract
Moderate to severe persistent pain after breast cancer surgery (PPBCS) affects 10 to 20% of the patients. Sensory dysfunction is often concomitantly present suggesting a neuropathic pain state. The relationship between various postoperative pain states and sensory dysfunction has been examined using quantitative sensory testing (QST), but only 2 smaller studies have examined PPBCS and sensory dysfunction in the surgical area. The purpose of this prospective study was to assess the relative importance of sensory function and PPBCS. QST consisted of sensory mapping, tactile detection threshold, mechanical pain threshold, and thermal thresholds. Two hundred ninety patients were enrolled and results showed that 38 (13%) had moderate to severe pain and 246 (85%) had hypoesthesia in the surgical area 1 year after surgery. Increased hypoesthesia areas were associated with pain at rest as well as during movement (P = .0001). Pain during movement was associated with a side-to-side difference of 140% (P = .001) for tactile detection threshold and 40% (P = .01) for mechanical pain threshold as well as increased thermal thresholds in the axilla (P > .001). Logistic regression models controlling for confounders showed larger areas of hypoesthesia as a significant risk factor, odds ratio 1.85 per 100 cm2 for pain at rest and odds ratio 1.36 per 100 cm2 for pain during movement. PERSPECTIVE PPBCS is associated with increasing areas of hypoesthesia as well as intraoperative nerve preservation. Thus, we hypothesize that PPBCS is associated with an interaction between a peripheral nociceptive drive in macroscopically preserved nerves and the central nervous system causing PPBCS as well as hypoesthesia. QST may identify patients suitable for intervention.
Collapse
Affiliation(s)
- Kenneth Geving Andersen
- Section for Surgical Pathophysiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Department of Breast Surgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | - Helle Molter Duriaud
- Section for Surgical Pathophysiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Kehlet
- Section for Surgical Pathophysiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Eske Kvanner Aasvang
- Section for Surgical Pathophysiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Anesthesiological Department, the Abdominal Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
142
|
Revisiting Mechanisms of Extraterritorial Allodynia. Curr Pain Headache Rep 2017; 21:25. [PMID: 28421380 DOI: 10.1007/s11916-017-0625-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
143
|
Breivik H, Stubhaug A, Butler S. CNS–mechanisms contribute to chronification of pain. Scand J Pain 2017; 15:137-139. [DOI: 10.1016/j.sjpain.2017.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Harald Breivik
- University of Oslo, Faculty of Medicine , Oslo , Norway
- Department of Anaesthesiology, Oslo University Hospital , Oslo , Norway
- Department of Pain Management and Research Oslo University Hospital , Oslo , Norway
| | - Audun Stubhaug
- University of Oslo, Faculty of Medicine , Oslo , Norway
- Department of Pain Management and Research Oslo University Hospital , Oslo , Norway
| | - Stephen Butler
- Multidisciplinary Pain Center, Academic Hospital , Uppsala , Sweden
- Department of Family and Preventive Medicine, Uppsala University , Uppsala , Sweden
| |
Collapse
|
144
|
Colloca L, Ludman T, Bouhassira D, Baron R, Dickenson AH, Yarnitsky D, Freeman R, Truini A, Attal N, Finnerup NB, Eccleston C, Kalso E, Bennett DL, Dworkin RH, Raja SN. Neuropathic pain. Nat Rev Dis Primers 2017; 3:17002. [PMID: 28205574 PMCID: PMC5371025 DOI: 10.1038/nrdp.2017.2] [Citation(s) in RCA: 1387] [Impact Index Per Article: 173.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neuropathic pain is caused by a lesion or disease of the somatosensory system, including peripheral fibres (Aβ, Aδ and C fibres) and central neurons, and affects 7-10% of the general population. Multiple causes of neuropathic pain have been described and its incidence is likely to increase owing to the ageing global population, increased incidence of diabetes mellitus and improved survival from cancer after chemotherapy. Indeed, imbalances between excitatory and inhibitory somatosensory signalling, alterations in ion channels and variability in the way that pain messages are modulated in the central nervous system all have been implicated in neuropathic pain. The burden of chronic neuropathic pain seems to be related to the complexity of neuropathic symptoms, poor outcomes and difficult treatment decisions. Importantly, quality of life is impaired in patients with neuropathic pain owing to increased drug prescriptions and visits to health care providers, as well as the morbidity from the pain itself and the inciting disease. Despite challenges, progress in the understanding of the pathophysiology of neuropathic pain is spurring the development of new diagnostic procedures and personalized interventions, which emphasize the need for a multidisciplinary approach to the management of neuropathic pain.
Collapse
Affiliation(s)
- Luana Colloca
- Department of Pain and Translational Symptom Science, School of Nursing and Department of Anesthesiology School of Medicine, University of Maryland, 655 West Lombard Street, 21201 Baltimore, Maryland, USA
| | - Taylor Ludman
- Department of Pain and Translational Symptom Science, School of Nursing and Department of Anesthesiology School of Medicine, University of Maryland, 655 West Lombard Street, 21201 Baltimore, Maryland, USA
| | - Didier Bouhassira
- INSERM, Unit 987, Ambroise Paré Hospital, UVSQ, Boulogne Billancourt, France
| | - Ralf Baron
- Department of Neurology, Division of Neurological Pain Research and Therapy, Klinik fur Neurologie Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Anthony H Dickenson
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - David Yarnitsky
- Department of Neurology, Rambam Health Care Campus, Technion Faculty of Medicine, Haifa, Israel
| | - Roy Freeman
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea Truini
- Department of Neurology and Psychiatry, Sapienza University, Rome, Italy
| | - Nadine Attal
- Pain Evaluation and Treatment Centre of Hôpital Ambroise Paré, Paris, France
| | - Nanna B Finnerup
- Department of Clinical Medicine - The Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - Christopher Eccleston
- Centre for Pain Research, University of Bath, Bath, UK
- Department of Clinical and Health Psychology, Ghent University, Ghent, Belgium
| | - Eija Kalso
- Division of Pain Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - David L Bennett
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Robert H Dworkin
- Department of Anesthesiology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
| | - Srinivasa N Raja
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
145
|
Poulsen JL, Olesen SS, Drewes AM, Ye B, Li WQ, Aghdassi AA, Sendler M, Mayerle J, Lerch MM. The Pathogenesis of Chronic Pancreatitis. CHRONIC PANCREATITIS 2017:29-62. [DOI: 10.1007/978-981-10-4515-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
146
|
|
147
|
Abstract
INTRODUCTION Recent advances regarding mechanisms of chronic pain emphasize the role of corticolimbic circuitry in predicting risk for chronic pain, independently from site of injury-related parameters. These results compel revisiting the role of peripheral nociceptive signaling in chronic pain. We address this issue by examining what brain circuitry transmit information regarding the intensity of chronic pain and how this information may be related to a common co-morbidity, depression. METHODS Resting state functional MRI was used in a large group of chronic pain patients (n=40 chronic back pain, CBP, and n=44 osteoarthritis, OA patients), and in comparison to healthy subjects (n=88). We used a graph theoretical measure, degree count, to investigate voxel-wise information sharing/transmission in the brain. Degree count, a functional connectivity based measure, identifies the number of voxels functionally connected to every given voxel. Subdividing the chronic pain cohort into discovery, replication, and also for overall group we show that only degree counts of diencephalic voxels centered in the ventral lateral thalamus reflected intensity of chronic pain, independently of depression. RESULTS Pain intensity was reliably associated with degree count of the thalamus, which was correlated negatively with components of the default mode network and positively with the periaqueductal grey (in contrast to healthy controls). Depression scores were not reliably associated with regional degree count. CONCLUSION Collectively the results suggest that, across two types of chronic pain, nociceptive specific information is relayed through the spinothalamic pathway to the lateral thalamus, potentiated by pro-nociceptive descending modulation, and interrupting cortical cognitive processes.
Collapse
|
148
|
|
149
|
Abstract
Traumatic nerve injuries can be devastating and life-changing events, leading to functional morbidity and psychological stress and social constraints. Even in the event of a successful surgical repair with recovered motor function, pain can result in continued disability and poor quality of life. Pain after nerve injury can also prevent recovery and return to preinjury life. It is difficult to predict which patients will develop persistent pain; once incurred, pain can be even challenging to manage. This review seeks to define the types of pain following peripheral nerve injuries, investigate the pathophysiology and causative factors, and evaluate potential treatment options.
Collapse
Affiliation(s)
- Gabrielle Davis
- Department of Surgery, Palo Alto VA, Suite 400, 770 Welch Road, Palo Alto, CA 94304, USA
| | - Catherine M Curtin
- Department of Surgery, Palo Alto VA, Suite 400, 770 Welch Road, Palo Alto, CA 94304, USA; Division of Plastic Surgery, Stanford University, Suite 400, 770 Welch Road, Palo Alto, CA 94304, USA.
| |
Collapse
|
150
|
Calvo M, Richards N, Schmid AB, Barroso A, Zhu L, Ivulic D, Zhu N, Anwandter P, Bhat MA, Court FA, McMahon SB, Bennett DLH. Altered potassium channel distribution and composition in myelinated axons suppresses hyperexcitability following injury. eLife 2016; 5:e12661. [PMID: 27033551 PMCID: PMC4841771 DOI: 10.7554/elife.12661] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/15/2016] [Indexed: 12/25/2022] Open
Abstract
Neuropathic pain following peripheral nerve injury is associated with hyperexcitability in damaged myelinated sensory axons, which begins to normalise over time. We investigated the composition and distribution of shaker-type-potassium channels (Kv1 channels) within the nodal complex of myelinated axons following injury. At the neuroma that forms after damage, expression of Kv1.1 and 1.2 (normally localised to the juxtaparanode) was markedly decreased. In contrast Kv1.4 and 1.6, which were hardly detectable in the naïve state, showed increased expression within juxtaparanodes and paranodes following injury, both in rats and humans. Within the dorsal root (a site remote from injury) we noted a redistribution of Kv1-channels towards the paranode. Blockade of Kv1 channels with α-DTX after injury reinstated hyperexcitability of A-fibre axons and enhanced mechanosensitivity. Changes in the molecular composition and distribution of axonal Kv1 channels, therefore represents a protective mechanism to suppress the hyperexcitability of myelinated sensory axons that follows nerve injury.
Collapse
Affiliation(s)
- Margarita Calvo
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom.,Departamento de Fisiologia, Facultad de Ciencias Biologicas- Pontificia Universidad Catolica de Chile, Santiago, Chile.,Departamento de Anestesiologia, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Natalie Richards
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom
| | - Annina B Schmid
- School of Health and Rehabilitation Sciences, The University of Queensland, Brisbane, Australia.,Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Alejandro Barroso
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom.,Hospital Regional Universitario de Málaga. Servicio de Anestesiología, Málaga, Spain
| | - Lan Zhu
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom.,School of Allied Health Sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, United Kingdom
| | - Dinka Ivulic
- Departamento de Fisiologia, Facultad de Ciencias Biologicas- Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Ning Zhu
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Philipp Anwandter
- Departamento Ortopedia y Traumatologia, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Manzoor A Bhat
- Department of Physiology, UT Health Science Center at San Antonio, San Antonio, United States.,School of Medicine, UT Health Science Center at San Antonio, San Antonio, United States
| | - Felipe A Court
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile.,FONDAP, Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Millenium Nucleus for Regenerative Biology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Stephen B McMahon
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom
| | - David L H Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|