1
|
Boehmerle W, Hagenacker T, Leo M, Schmitt LI, Lehmann HC, Klein I, Stegherr R, Konietschke F, Endres M, Huehnchen P. Results of the preclinical multicenter randomized controlled paclitaxel-induced neuropathy prevention replication study (PINPRICS). BMC Res Notes 2025; 18:145. [PMID: 40200318 PMCID: PMC11978143 DOI: 10.1186/s13104-025-07206-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
OBJECTIVE Chemotherapy-induced peripheral neuropathy (CIPN) is a frequent and serious side effect of many cytotoxic drugs, including paclitaxel. Despite the identification of treatment options in animal models, clinical trials for the treatment or prevention of CIPN have been negative. Major challenges for successful clinical translation of preclinical data include a lack of reproducibility and randomization, small sample sizes and insufficient statistical tests. We therefore conducted a confirmatory, preclinical multicenter randomized controlled replication trial to test the safety and efficacy of three drugs for preventing paclitaxel-induced polyneuropathy: (1) nilotinib, (2) lithium carbonate and (3) interleukin-6-neutralizing antibodies. We preregistered the data analysis plan as well as the two-step study protocol: the optimal doses of the three compounds were assessed first and then tested in a mouse breast cancer xenograft model to compare safety and efficacy. RESULTS Unfortunately, toxicity of intraperitoneally administered nilotinib in combination with paclitaxel was observed, and higher-than-expected tumor growth resulted in a lack of power when the trial was analyzed. Thus, although lithium carbonate and IL-6-neutralizing antibodies tended toward neuroprotection, the differences between these groups were not statistically significant. However, the PINPRICS study ultimately still provides important lessons with regard to the planning and conduction of multicenter preclinical trials.
Collapse
Affiliation(s)
- Wolfgang Boehmerle
- Klinik und Hochschulambulanz für Neurologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117, Berlin, Germany.
| | - Tim Hagenacker
- Department of Neurology and Center for Translational Neuro- and Behavioral Science, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Markus Leo
- Department of Neurology and Center for Translational Neuro- and Behavioral Science, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Linda-Isabell Schmitt
- Department of Neurology and Center for Translational Neuro- and Behavioral Science, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Helmar C Lehmann
- Department of Neurology, Medical Faculty, University Hospital Cologne, 50937, Köln, Germany
| | - Ines Klein
- Department of Neurology, Medical Faculty, University Hospital Cologne, 50937, Köln, Germany
| | - Regina Stegherr
- Institut für Biometrie und Klinische Epidemiologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117, Berlin, Germany
| | - Frank Konietschke
- Institut für Biometrie und Klinische Epidemiologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117, Berlin, Germany
| | - Matthias Endres
- Klinik und Hochschulambulanz für Neurologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117, Berlin, Germany
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Partner site, Berlin, Germany
- Partner site, German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Partner site, German Center for Mental Health (DZPG), Berlin, Germany
| | - Petra Huehnchen
- Klinik und Hochschulambulanz für Neurologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
2
|
Pangeni R, Poudel S, Herz SM, Berkbigler G, Duerfeldt AS, Damaj MI, Xu Q. New PPARα Agonist A190-Loaded Microemulsion for Chemotherapy-Induced Peripheral Neuropathy. Mol Pharm 2025; 22:1641-1656. [PMID: 39879378 PMCID: PMC11881135 DOI: 10.1021/acs.molpharmaceut.4c01374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a serious side effect of anticancer agents with limited effective preventive or therapeutic interventions. Although fenofibrate, a peroxisome proliferator-activated receptor-alpha (PPARα) agonist, has demonstrated neuroprotective and analgesic properties, its clinical utility is hindered by low receptor affinity, poor subtype selectivity, and suboptimal bioavailability. A190, a highly selective and potent nonfibrate PPARα agonist, offers a promising alternative but is limited by poor aqueous solubility, resulting in reduced oral bioavailability and therapeutic efficacy. To address these limitations, an optimized oil-in-water (o/w) microemulsion formulation was developed using Box-Behnken design to enhance the solubility and intestinal permeability of A190. The A190 microemulsion exhibited physical stability with a droplet size of approximately 100 nm and a drug loading efficiency of greater than 95%. The effective and apparent permeability of A190 from the microemulsion was significantly higher compared to that of free A190 dispersion, respectively. Additionally, no significant impact on the cell viability was observed, indicating less toxicity and a good biocompatibility of the formulation components. The oral bioavailability of A190 microemulsion was approximately 5-fold higher compared to A190 dispersion, demonstrating the microemulsion's potential to greatly enhance the oral bioavailability of hydrophobic drugs. Furthermore, our findings reveal that orally administered A190 microemulsion effectively reduced CIPN-induced mechanical hypersensitivity, likely mediated through PPARα activation. A190 microemulsion was found to be equally effective at reducing the chronic inflammatory complete Freund's adjuvant-induced pain. These results underscore A190s potential as a nonopioid therapeutic candidate, utilizing a novel microemulsion formulation for the management of chemotherapy-induced neuropathic pain and chronic inflammatory pain.
Collapse
Affiliation(s)
- Rudra Pangeni
- Department
of Pharmaceutics, School of Pharmacy, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
| | - Surendra Poudel
- Department
of Pharmaceutics, School of Pharmacy, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
| | - Sara M. Herz
- Department
of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Grant Berkbigler
- Department
of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Adam S. Duerfeldt
- Department
of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - M. Imad Damaj
- Department
of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Qingguo Xu
- Department
of Pharmaceutics, School of Pharmacy, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
- Departments
of Ophthalmology, Pediatrics, Biomedical Engineering, and Massey Cancer
Center, Center for Pharmaceutical Engineering, and Center for Drug
Discovery, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
3
|
Huerta MÁ, Cisneros E, Alique M, Roza C. Strategies for measuring non-evoked pain in preclinical models of neuropathic pain: Systematic review. Neurosci Biobehav Rev 2024; 163:105761. [PMID: 38852847 DOI: 10.1016/j.neubiorev.2024.105761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
The development of new analgesics for neuropathic pain treatment is crucial. The failure of promising drugs in clinical trials may be related to the over-reliance on reflex-based responses (evoked pain) in preclinical drug testing, which may not fully represent clinical neuropathic pain, characterized by spontaneous non-evoked pain (NEP). Hence, strategies for assessing NEP in preclinical studies emerged. This systematic review identified 443 articles evaluating NEP in neuropathic pain models (mainly traumatic nerve injuries in male rodents). An exponential growth in NEP evaluation was observed, which was assessed using 48 different tests classified in 12 NEP-related outcomes: anxiety, exploration/locomotion, paw lifting, depression, conditioned place preference, gait, autotomy, wellbeing, facial grooming, cognitive impairment, facial pain expressions and vocalizations. Although most of these outcomes showed clear limitations, our analysis suggests that conditioning-associated outcomes, pain-related comorbidities, and gait evaluation may be the most effective strategies. Moreover, a minimal part of the studies evaluated standard analgesics. The greater emphasis on evaluating NEP aligning with clinical pain symptoms may enhance analgesic drug development, improving clinical translation.
Collapse
Affiliation(s)
- Miguel Á Huerta
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada 18016, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, 18100 Armilla, Granada, Spain; Biosanitary Research Institute ibs.GRANADA, Granada 18012, Spain
| | - Elsa Cisneros
- Health Sciences School, Centro Universitario Internacional de Madrid (CUNIMAD), Madrid, Spain; Health Sciences School, Universidad Internacional de La Rioja (UNIR), Logroño, Spain
| | - Matilde Alique
- Department of System's Biology, Medical School, University of Alcala de Henares, Alcalá de Henares, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Carolina Roza
- Department of System's Biology, Medical School, University of Alcala de Henares, Alcalá de Henares, Spain.
| |
Collapse
|
4
|
Bakare AO, Stephens K, Sanchez KR, Liu V, Zheng L, Goel V, Guan Y, Sivanesan E. Spinal cord stimulation attenuates paclitaxel-induced gait impairment and mechanical hypersensitivity via peripheral neuroprotective mechanisms in tumor-bearing rats. Reg Anesth Pain Med 2024:rapm-2024-105433. [PMID: 38844412 PMCID: PMC11645439 DOI: 10.1136/rapm-2024-105433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Taxanes such as paclitaxel (PTX) induce dose-dependent chemotherapy-induced peripheral neuropathy (CIPN), which is associated with debilitating chronic pain and gait impairment. Increased macrophage-related proinflammatory activities have been reported to mediate the development and maintenance of neuropathic pain. While spinal cord stimulation (SCS) has been used for a number of pain conditions, the mechanisms supporting its use for CIPN remain to be elucidated. Thus, we aimed to examine whether SCS can attenuate Schwann cell-mediated and macrophage-mediated neuroinflammation in the sciatic nerve of Rowlette Nude (RNU) rats with PTX-induced gait impairment and mechanical hypersensitivity. METHODS Adult male tumor-bearing RNU rats were used for this study examining PTX treatment and SCS. Gait and mechanical hypersensitivity were assessed weekly. Cytokines, gene expression, macrophage infiltration and polarization, nerve morphology and Schwann cells were examined in sciatic nerves using multiplex immunoassay, bulk RNA sequencing, histochemistry and immunohistochemistry techniques. RESULTS SCS (50 Hz, 0.2 milliseconds, 80% motor threshold) attenuated the development of mechanical hypersensitivity (20.93±0.80 vs 12.23±2.71 grams, p<0.0096) and temporal gait impairment [swing (90.41±7.03 vs 117.27±9.71%, p<0.0076), and single stance times (94.92±3.62 vs 112.75±7.27%, p<0.0245)] induced by PTX (SCS+PTX+Tumor vs Sham SCS+PTX+Tumor). SCS also attenuated the reduction in Schwann cells, myelin thickness and increased the concentration of anti-inflammatory cytokine interleukin (IL)-10. Bulk RNA sequencing revealed differential gene expression after SCS, with 607 (59.2%) genes upregulated while 418 (40.8%) genes were downregulated. Notably, genes related to anti-inflammatory cytokines and neuronal growth were upregulated, while genes related to proinflammatory-promoting genes, increased M2γ polarization and decreased macrophage infiltration and Schwann cell loss were downregulated. CONCLUSION SCS may attenuate PTX-induced pain and temporal gait impairment, which may be partly attributed to decreases in Schwann cell loss and macrophage-mediated neuroinflammation in sciatic nerves.
Collapse
Affiliation(s)
- Ahmed Olalekan Bakare
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kimberly Stephens
- Arkansas Children's Research Institute, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Karla R Sanchez
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vivian Liu
- Department of Computer Science, Johns Hopkins Whiting School of Engineering, Baltimore, Maryland, USA
| | - Lei Zheng
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vasudha Goel
- Department of Anesthesiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurological Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eellan Sivanesan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Lucarini E, Micheli L, Rajagopalan R, Ciampi C, Branca JJ, Pacini A, Leandri M, Rajagopalan P, Ghelardini C, Di Cesare Mannelli L. Broad-spectrum neuroprotection exerted by DDD-028 in a mouse model of chemotherapy-induced neuropathy. Pain 2023; 164:2581-2595. [PMID: 37556385 PMCID: PMC10578426 DOI: 10.1097/j.pain.0000000000002963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/28/2023] [Accepted: 05/02/2023] [Indexed: 08/11/2023]
Abstract
ABSTRACT Neurotoxicity of chemotherapeutics involves peculiar alterations in the structure and function, including abnormal nerve signal transmission, of both the peripheral and central nervous system. The lack of effective pharmacological approaches to prevent chemotherapy-induced neurotoxicity necessitates the identification of innovative therapies. Recent evidence suggests that repeated treatment with the pentacyclic pyridoindole derivative DDD-028 can exert both pain-relieving and glial modulatory effects in mice with paclitaxel-induced neuropathy. This work is aimed at assessing whether DDD-028 is a disease-modifying agent by protecting the peripheral nervous tissues from chemotherapy-induced damage. Neuropathy was induced in animals by paclitaxel injection (2.0 mg kg -1 i.p). DDD-028 (10 mg kg -1 ) and the reference drug, pregabalin (30 mg kg -1 ), were administered per os daily starting concomitantly with the first injection of paclitaxel and continuing 10 days after the end of paclitaxel treatment. The behavioural tests confirmed the antihyperalgesic efficacy of DDD-028 on paclitaxel-induced neuropathic pain. Furthermore, the electrophysiological analysis revealed the capacity of DDD-028 to restore near-normal sensory nerve conduction in paclitaxel-treated animals. Histopathology evidence indicated that DDD-028 was able to counteract effectively paclitaxel-induced peripheral neurotoxicity by protecting against the loss of intraepidermal nerve fibers, restoring physiological levels of neurofilament in nerve tissue and plasma, and preventing morphological alterations occurring in the sciatic nerves and dorsal root ganglia. Overall, DDD-028 is more effective than pregabalin in preventing chemotherapy-induced neurotoxicity. Thus, based on its potent antihyperalgesic and neuroprotective efficacy, DDD-028 seems to be a viable prophylactic medication to limit the development of neuropathies consequent to chemotherapy.
Collapse
Affiliation(s)
- Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | | | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Jacopo J.V. Branca
- Department of Experimental and Clinical Medicine, Anatomy and Histology Section, University of Florence, Florence, Italy
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine, Anatomy and Histology Section, University of Florence, Florence, Italy
| | - Massimo Leandri
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | | | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| |
Collapse
|
6
|
Lopez-Garzon M, Canta A, Chiorazzi A, Alberti P. Gait analysis in chemotherapy-induced peripheral neurotoxicity rodent models. Brain Res Bull 2023; 203:110769. [PMID: 37748696 DOI: 10.1016/j.brainresbull.2023.110769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/05/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023]
Abstract
Gait analysis could be used in animal models as an indicator of sensory ataxia due to chemotherapy-induced peripheral neurotoxicity (CIPN). Over the years, gait analysis in in vivo studies has evolved from simple observations carried out by a trained operator to computerised systems with machine learning that allow the quantification of any variable of interest and the establishment of algorithms for behavioural classification. However, there is not a consensus on gait analysis use in CIPN animal models; therefore, we carried out a systematic review. Of 987 potentially relevant studies, 14 were included, in which different methods were analysed (observation, footprint and CatWalk™). We presented the state-of-the-art of possible approaches to analyse sensory ataxia in rodent models, addressing advantages and disadvantages of different methods available. Semi-automated methods may be of interest when preventive or therapeutic strategies are evaluated, also considering their methodological simplicity and automaticity; up to now, only CatWalk™ analysis has been tested. Future studies should expect that CIPN-affected animals tend to reduce hind paw support due to pain, allodynia or loss of sensation, and an increase in swing phase could or should be observed. Few available studies documented these impairments at the last time point, and only appeared later on respect to other earlier signs of CIPN (such as altered neurophysiological findings). For that reason, gait impairment could be interpreted as late repercussions of loss of sensory.
Collapse
Affiliation(s)
- Maria Lopez-Garzon
- Biomedical Group (BIO277), Department of Physiotherapy, Faculty of Health Sciences, University of Granada, Granada, Spain; A02-Cuídate, Instituto de Investigación Biosanitaria Ibs, GRANADA, Granada, Spain; Unit of Excellence On Exercise and Health (UCEES), University of Granada, Granada, Spain; Sport and Health Research Center (IMUDs), Granada, Spain
| | - Annalisa Canta
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy; NeuroMI (Milan Center for neuroscience), Milan, Italy
| | - Alessia Chiorazzi
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy; NeuroMI (Milan Center for neuroscience), Milan, Italy
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy; NeuroMI (Milan Center for neuroscience), Milan, Italy; Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
| |
Collapse
|
7
|
Klazas M, Naamneh MS, Zheng W, Lazarovici P. Gabapentin Increases Intra-Epidermal and Peptidergic Nerve Fibers Density and Alleviates Allodynia and Thermal Hyperalgesia in a Mouse Model of Acute Taxol-Induced Peripheral Neuropathy. Biomedicines 2022; 10:biomedicines10123190. [PMID: 36551946 PMCID: PMC9775678 DOI: 10.3390/biomedicines10123190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
The clinical pathology of Taxol-induced peripheral neuropathy (TIPN), characterized by loss of sensory sensitivity and pain, is mirrored in a preclinical pharmacological mice model in which Gabapentin, produced anti-thermal hyperalgesia and anti-allodynia effects. The study aimed to investigate the hypothesis that gabapentin may protect against Taxol-induced neuropathic pain in association with an effect on intra-epidermal nerve fibers density in the TIPN mice model. A TIPN study schedule was induced in mice by daily injection of Taxol during the first week of the experiment. Gabapentin therapy was performed during the 2nd and 3rd weeks. The neuropathic pain was evaluated during the whole experiment by the Von Frey, tail flick, and hot plate tests. Intra-epidermal nerve fibers (IENF) density in skin biopsies was measured at the end of the experiment by immunohistochemistry of ubiquitin carboxyl-terminal hydrolase PGP9.5 pan-neuronal and calcitonin gene-related (CGRP) peptides-I/II- peptidergic markers. Taxol-induced neuropathy was expressed by 80% and 73% reduction in the paw density of IENFs and CGPR, and gabapentin treatment corrected by 83% and 46% this reduction, respectively. Gabapentin-induced increase in the IENF and CGRP nerve fibers density, thus proposing these evaluations as an additional objective end-point tool in TIPN model studies using gabapentin as a reference compound.
Collapse
Affiliation(s)
- Michal Klazas
- Pharmacy Unit, School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Majdi Saleem Naamneh
- Pharmacology Unit, School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Wenhua Zheng
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Philip Lazarovici
- Pharmacology Unit, School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112002, Israel
- Correspondence: ; Tel.: +972-2-6758729; Fax: +972-2-6757490
| |
Collapse
|
8
|
Dallazen JL, da Luz BB, Maria-Ferreira D, Nascimento AM, Cipriani TR, de Souza LM, Geppetti P, de Paula Werner MF. Local effects of natural alkylamides from Acmella oleracea and synthetic isobutylalkyl amide on neuropathic and postoperative pain models in mice. Fitoterapia 2022; 160:105224. [PMID: 35659524 DOI: 10.1016/j.fitote.2022.105224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 11/25/2022]
Abstract
Neuropathic and postoperative pain are clinical conditions that impair the patient's quality of life. The current pharmacotherapy of both painful states is ineffective and accompanied by several side effects. In order to develop new therapeutics targets, the secondary metabolites of plants have been extensively studied. Acmella oleracea ("jambu") is a native plant from the Amazon region and rich in alkylamides, bioactive compounds responsible for inducing anesthetic and chemesthetic sensations. We previously demonstrated that the intraplantar administration of an hexanic fraction (HF) rich in alkylamides from jambu and the synthetic isobutylalkyl amide (IBA) at 0.1 μg/20 μL can promote antinociceptive and anti-inflammatory effects. Thus, this study aimed to evaluate the local effect of HF and IBA (0.1 μg/20 μL) on neuropathic (partial sciatic nerve ligation, PSNL) and postoperative pain (plantar incision surgery, PIS) models in mice. Seven days after the PSNL, the mechanical (von Frey test) and cold (acetone-evoked evaporative cooling) allodynia, and digital gait parameters were analyzed. The intraplantar HF and IBA treatments attenuated the mechanical and cold allodynia as well as the static (max. Contact and print area) and dynamic (stand duration) parameters of digital gait analyses. On the day after PIS, the mechanical allodynia, heat hyperalgesia (hot plate, 52 ± 0.1°C), and spontaneous nociception scores were evaluated. Topical treatment with HF reduced the mechanical allodynia, heat hyperalgesia, and spontaneous nociception scores. In contrast, IBA treatment only partially reduced the mechanical allodynia. In summary, the local treatment with HF was effective on both neuropathic and postoperative pain, as opposed to IBA, which only had an effect on neuropathic pain.
Collapse
Affiliation(s)
| | | | - Daniele Maria-Ferreira
- Department of Pharmacology, Federal University of Parana, Curitiba, Brazil; Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Adamara Machado Nascimento
- Department of Biochemistry and Molecular Biology, Federal University of Parana, Curitiba, Brazil; Multidisciplinary Center, Federal University of Acre, Cruzeiro do Sul, Brazil
| | - Thales Ricardo Cipriani
- Department of Biochemistry and Molecular Biology, Federal University of Parana, Curitiba, Brazil
| | - Lauro Mera de Souza
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Pierangelo Geppetti
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | | |
Collapse
|
9
|
Caudle RM, Neubert JK. Effects of Oxaliplatin on Facial Sensitivity to Cool Temperatures and TRPM8 Expressing Trigeminal Ganglion Neurons in Mice. FRONTIERS IN PAIN RESEARCH 2022; 3:868547. [PMID: 35634452 PMCID: PMC9130462 DOI: 10.3389/fpain.2022.868547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/18/2022] [Indexed: 12/04/2022] Open
Abstract
The chemotherapeutic agent oxaliplatin is commonly used to treat colorectal cancer. Although effective as a chemotherapeutic, it frequently produces painful peripheral neuropathies. These neuropathies can be divided into an acute sensitivity to cool temperatures in the mouth and face, and chronic neuropathic pain in the limbs and possible numbness. The chronic neuropathy also includes sensitivity to cool temperatures. Neurons that detect cool temperatures are reported to utilize Transient Receptor Potential Cation Channel, Subfamily M, Member 8 (TRPM8). Therefore, we investigated the effects of oxaliplatin on facial nociception to cool temperatures (18°C) in mice and on TRPM8 expressing trigeminal ganglion (TRG) neurons. Paclitaxel, a chemotherapeutic that is used to treat breast cancer, was included for comparison because it produces neuropathies, but acute cool temperature sensitivity in the oral cavity or face is not typically reported. Behavioral testing of facial sensitivity to 18°C indicated no hypersensitivity either acutely or chronically following either chemotherapeutic agent. However, whole cell voltage clamp experiments in TRPM8 expressing TRG neurons indicated that both oxaliplatin and paclitaxel increased Hyperpolarization-Activated Cyclic Nucleotide-Gated channel (HCN), voltage gated sodium channel (Nav), and menthol evoked TRPM8 currents. Voltage gated potassium channel (Kv) currents were not altered. Histological examination of TRPM8 fibers in the skin of the whisker pads demonstrated that the TRPM8 expressing axons and possible Merkel cell-neurite complexes were damaged by oxaliplatin. These findings indicate that oxaliplatin induces a rapid degeneration of TRG neuron axons that express TRPM8, which prevents evoked activation of the sensitized neurons and likely leads to reduced sensitivity to touch and cool temperatures. The changes in HCN, Nav, and TRPM8 currents suggest that spontaneous firing of action potentials may be increased in the deafferented neurons within the ganglion, possibly producing spontaneously induced cooling or nociceptive sensations.
Collapse
Affiliation(s)
- Robert M. Caudle
- Department of Oral and Maxillofacial Surgery, University of Florida, Gainesville, FL, United States
| | - John K. Neubert
- Department of Orthodontics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
10
|
Livni L, Keating BA, Fiore NT, Lees JG, Goldstein D, Moalem-Taylor G. Effects of combined chemotherapy and anti-programmed cell death protein 1 treatment on peripheral neuropathy and neuroinflammation in mice. Pain 2022; 163:110-124. [PMID: 34224494 DOI: 10.1097/j.pain.0000000000002384] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/27/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT A modern approach for cancer treatment is the use of immunotherapy, and particularly immune checkpoint inhibitors, such as anti-programmed cell death protein 1 (PD-1), alone and in combination with chemotherapy. The PD-1 pathway plays a crucial role in inhibiting immune responses and recently has been shown to modulate neuronal activity. However, the impact of PD-1 blockade on the development of chemotherapy-induced peripheral neuropathy is currently unknown. In this study, we show that C57BL/6 mice treated with the chemotherapeutic drug paclitaxel or cotherapy (paclitaxel and anti-PD-1), but not with anti-PD-1 alone, exhibited increased mechanical sensitivity of the hind paw. Both chemotherapy and immunotherapy caused a reduction in neurite outgrowth of dorsal root ganglion (DRG) explants derived from treated mice, whereas only paclitaxel reduced the neurite outgrowth after direct in vitro treatment. Mice treated with anti-PD-1 or cotherapy exhibited distinct T-cell changes in the lymph nodes and increased T-cell infiltration into the DRG. Mice treated with paclitaxel or cotherapy had increased macrophage presence in the DRG, and all treated groups presented an altered expression of microglia markers in the dorsal horn of the spinal cord. We conclude that combining anti-PD-1 immunotherapy with paclitaxel does not increase the severity of paclitaxel-induced peripheral neuropathy. However, because anti-PD-1 treatment caused significant changes in DRG and spinal cord immunity, caution is warranted when considering immune checkpoint inhibitors therapy in patients with a high risk of developing neuropathy.
Collapse
Affiliation(s)
- Lital Livni
- Department of Physiology, Translational Neuroscience Facility, School of Medical Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Brooke A Keating
- Department of Physiology, Translational Neuroscience Facility, School of Medical Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Nathan T Fiore
- Department of Physiology, Translational Neuroscience Facility, School of Medical Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Justin G Lees
- Department of Physiology, Translational Neuroscience Facility, School of Medical Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - David Goldstein
- Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia
- Department of Medical Oncology, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Gila Moalem-Taylor
- Department of Physiology, Translational Neuroscience Facility, School of Medical Sciences, The University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
11
|
Kang DW, Choi JG, Kim J, Park JB, Lee JH, Kim HW. Bee venom reduces burn-induced pain via the suppression of peripheral and central substance P expression in mice. J Vet Sci 2021; 22:e9. [PMID: 33522161 PMCID: PMC7850793 DOI: 10.4142/jvs.2021.22.e9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/03/2020] [Accepted: 12/13/2020] [Indexed: 11/20/2022] Open
Abstract
Background Scalding burn injuries can occur in everyday life but occur more frequently in young children. Therefore, it is important to develop more effective burn treatments. Objectives This study examined the effects of bee venom (BV) stimulation on scalding burn injury-induced nociception in mice as a new treatment for burn pain. Methods To develop a burn injury model, the right hind paw was immersed temporarily in hot water (65°C, 3 seconds). Immediately after the burn, BV (0.01, 0.02, or 0.1 mg/kg) was injected subcutaneously into the ipsilateral knee area once daily for 14 days. A von Frey test was performed to assess the nociceptive response, and the altered walking parameters were evaluated using an automated gait analysis system. In addition, the peripheral and central expression changes in substance P (Sub P) were measured in the dorsal root ganglion and spinal cord by immunofluorescence. Results Repeated BV treatment at the 2 higher doses used in this study (0.02 and 0.1 mg/kg) alleviated the pain responses remarkably and recovered the gait performances to the level of acetaminophen (200 mg/kg, intraperitoneal, once daily), which used as the positive control group. Moreover, BV stimulation had an inhibitory effect on the increased expression of Sub P in the peripheral and central nervous systems by a burn injury. Conclusions These results suggest that a peripheral BV treatment may have positive potency in treating burn-induced pain.
Collapse
Affiliation(s)
- Dong Wook Kang
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon 35015, Korea
| | - Jae Gyun Choi
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon 35015, Korea
| | - Jaehyuk Kim
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon 35015, Korea
| | - Jin Bong Park
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon 35015, Korea
| | - Jang Hern Lee
- Department of Veterinary Physiology, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Hyun Woo Kim
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon 35015, Korea.
| |
Collapse
|
12
|
Jelgersma C, Vajkoczy P. How to Target Spinal Metastasis in Experimental Research: An Overview of Currently Used Experimental Mouse Models and Future Prospects. Int J Mol Sci 2021; 22:ijms22115420. [PMID: 34063821 PMCID: PMC8196562 DOI: 10.3390/ijms22115420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/12/2021] [Accepted: 05/18/2021] [Indexed: 01/20/2023] Open
Abstract
The spine is one of the organs that is most affected by metastasis in cancer patients. Since the control of primary tumor is continuously improving, treatment of metastases is becoming one of the major challenges to prevent cancer-related death. Due to the anatomical proximity to the spinal cord, local spread of metastasis can directly cause neurological deficits, severely limiting the patient’s quality of life. To investigate the underlying mechanisms and to develop new therapies, preclinical models are required which represent the complexity of the multistep cascade of metastasis. Current research of metastasis focuses on the formation of the premetastatic niche, tumor cell dormancy and the influence and regulating function of the immune system. To unveil whether these influence the organotropism to the spine, spinal models are irreplaceable. Mouse models are one of the most suitable models in oncologic research. Therefore, this review provides an overview of currently used mouse models of spinal metastasis. Furthermore, it discusses technical aspects clarifying to what extend these models can picture key steps of the metastatic process. Finally, it addresses proposals to develop better mouse models in the future and could serve as both basis and stimulus for researchers and clinicians working in this field.
Collapse
|
13
|
Bluette CT, Shoieb AM, Peng Q, Manickam B, Huang W, Shin E, Zhang W, Song YH, Liu CN. Behavioral, Histopathologic, and Molecular Biological Responses of Nanoparticle- and Solution-Based Formulations of Vincristine in Mice. Int J Toxicol 2020; 40:40-51. [PMID: 33148080 DOI: 10.1177/1091581820968255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Clinical use of the chemotherapeutic agent vincristine (VCR) is limited by chemotherapy-induced peripheral neuropathy (CiPN). A new formulation of VCR encapsulated by nanoparticles has been proposed and developed to alleviate CiPN. We hypothesized in nonclinical animals that the nanoparticle drug would be less neurotoxic due to different absorption and distribution properties to the peripheral nerve from the unencapsulated free drug. Here, we assessed whether VCR encapsulation in nanoparticles alleviates CiPN using behavioral gait analysis (CatWalk), histopathologic and molecular biological (RT-qPCR) approaches. Adult male C57BL/6 mice were assigned to 3 groups (empty nanoparticle, nano-VCR, solution-based VCR, each n = 8). After 15 days of dosing, animals were euthanized for tissue collection. It was shown that intraperitoneal administration of nano-VCR (0.15 mg/kg, every other day) and the empty nanoparticle resulted in no changes in gait parameters; whereas, injection of solution-based VCR resulted in decreased run speed and increased step cycle and stance (P < 0.05). There were no differences in incidence and severity of degeneration in the sciatic nerves between the nano-VCR-dosed and solution-based VCR-dosed animals. Likewise, decreased levels of a nervous tissue-enriched microRNA-183 in circulating blood did not show a significant difference between the nano- and solution-based VCR groups (P > 0.05). Empty nanoparticle administration did not cause any behavioral, microRNA, or structural changes. In conclusion, this study suggests that the nano-VCR formulation may alleviate behavioral changes in CiPN, but it does not improve the structural changes of CiPN in peripheral nerve. Nanoparticle properties may need to be optimized to improve biological observations.
Collapse
Affiliation(s)
- Crystal T Bluette
- Comparative Medicine, 105623Pfizer Worldwide RD&M, Cambridge, MA, USA
| | - Ahmed M Shoieb
- Drug Safety R&D, 105623Pfizer Worldwide RD&M, Groton, CT, USA
| | - Qinghai Peng
- Drug Safety R&D, 105623Pfizer Worldwide RD&M, San Diego, CA, USA
| | | | - Wenhu Huang
- Drug Safety R&D, 105623Pfizer Worldwide RD&M, San Diego, CA, USA
| | - Eyoung Shin
- Oncology, 105623Pfizer Worldwide RD&M, Pearl River, NY, USA
| | - Wei Zhang
- Oncology, 105623Pfizer Worldwide RD&M, Pearl River, NY, USA
| | - Young-Ho Song
- Oncology, 105623Pfizer Worldwide RD&M, Pearl River, NY, USA
| | - Chang-Ning Liu
- Comparative Medicine, 105623Pfizer Worldwide RD&M, Groton, CT, USA. Peng is now with Protego Biopharma, San Diego, CA, USA
| |
Collapse
|
14
|
Heinzel J, Längle G, Oberhauser V, Hausner T, Kolbenschlag J, Prahm C, Grillari J, Hercher D. Use of the CatWalk gait analysis system to assess functional recovery in rodent models of peripheral nerve injury - a systematic review. J Neurosci Methods 2020; 345:108889. [PMID: 32755615 DOI: 10.1016/j.jneumeth.2020.108889] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
Injuries of the peripheral nervous system are common among the population affecting around 3% of all trauma patients. This high clinical need in the field of peripheral nerve injury and regeneration has been steadily driving experimental and epidemiological research. Thereby, it is crucial to determine the exact degree of recovery of end-organ function. Regeneration after nerve injuries is assessed by a wide variety of techniques and pre-clinical model systems, where rodent models are among the most widely used. However, results from rodents are difficult to translate to human patients in general, and reproducible and comparable assessment of functional recovery is of highest importance. Computerized gait analysis allows comprehensive acquisition of locomotor function. As the animals cross the recording device voluntarily, functional recovery is assessable with a minimum degree of human interference on their behavior. This article aims to give a detailed overview on the existing literature on CatWalk gait analysis in rodent models of peripheral nerve injuries of upper and lower extremities, e.g. axonotmesis, neurotmesis or fibrosis, with special emphasis on differences between models. Researchers interested in assessment of locomotor function in such models will especially benefit from this work as it will provide them with an overview of the various experimental setups and expected outcomes. This work also addresses potential pitfalls and hurdles in order to promote well designed, comparable studies allowing for accelerated development of therapeutic strategies in peripheral repair and regeneration.
Collapse
Affiliation(s)
- Johannes Heinzel
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Trauma Center Tuebingen, Eberhard Karls University, Tuebingen, Germany; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, 1020, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Gregor Längle
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, 1020, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Viola Oberhauser
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, 1020, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Thomas Hausner
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, 1020, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Jonas Kolbenschlag
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Trauma Center Tuebingen, Eberhard Karls University, Tuebingen, Germany
| | - Cosima Prahm
- Department of Hand-, Plastic, Reconstructive and Burn Surgery, BG Trauma Center Tuebingen, Eberhard Karls University, Tuebingen, Germany
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, 1020, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria; Institute of Molecular Biotechnology, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - David Hercher
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstraße 13, 1020, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria.
| |
Collapse
|
15
|
Kiven S, Wang Y, Aich A, Argueta DA, Lei J, Sagi V, Tennakoon M, Bedros SJ, Lambrecht N, Gupta K. Spatiotemporal Alterations in Gait in Humanized Transgenic Sickle Mice. Front Immunol 2020; 11:561947. [PMID: 33178189 PMCID: PMC7593487 DOI: 10.3389/fimmu.2020.561947] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022] Open
Abstract
Sickle cell disease (SCD) is a hemoglobinopathy affecting multiple organs and featuring acute and chronic pain. Purkinje cell damage and hyperalgesia have been demonstrated in transgenic sickle mice. Purkinje cells are associated with movement and neural function which may influence pain. We hypothesized that Purkinje cell damage and/or chronic pain burden provoke compensatory gait changes in sickle mice. We found that Purkinje cells undergoe increased apoptosis as shown by caspase-3 activation. Using an automated gait measurement system, MouseWalker, we characterized spatiotemporal gait characteristics of humanized transgenic BERK sickle mice in comparison to control mice. Sickle mice showed alteration in stance instability and dynamic gait parameters (walking speed, stance duration, swing duration and specific swing indices). Differences in stance instability may reflect motor dysfunction due to damaged Purkinje cells. Alterations in diagonal and all stance indices indicative of hesitation during walking may originate from motor dysfunction and/or arise from fear and/or anticipation of movement-evoked pain. We also demonstrate that stance duration, diagonal swing indices and all stance indices correlate with both mechanical and deep tissue hyperalgesia, while stance instability correlates with only deep tissue hyperalgesia. Therefore, objective analysis of gait in SCD may provide insights into neurological impairment and pain states.
Collapse
Affiliation(s)
- Stacy Kiven
- Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN, United States
| | - Ying Wang
- Department of Anesthesia, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Anupam Aich
- Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Donovan A. Argueta
- Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Jianxun Lei
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN, United States
| | - Varun Sagi
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN, United States
| | - Madhushan Tennakoon
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN, United States
| | - Saad J. Bedros
- College of Science & Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Nils Lambrecht
- Pathology and Laboratory Medicine, Long Beach VA Healthcare System, Long Beach, CA, United States
| | - Kalpna Gupta
- Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN, United States
- Southern California Institute for Research and Education, Long Beach VA Healthcare System, Long Beach, CA, United States
| |
Collapse
|
16
|
Broggini T, Piffko A, Hoffmann CJ, Ghori A, Harms C, Adams RH, Vajkoczy P, Czabanka M. Ephrin-B2-EphB4 communication mediates tumor-endothelial cell interactions during hematogenous spread to spinal bone in a melanoma metastasis model. Oncogene 2020; 39:7063-7075. [PMID: 32989254 DOI: 10.1038/s41388-020-01473-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 08/21/2020] [Accepted: 09/15/2020] [Indexed: 11/09/2022]
Abstract
Metastases account for the majority of cancer deaths. Bone represents one of the most common sites of distant metastases, and spinal bone metastasis is the most common source of neurological morbidity in cancer patients. During metastatic seeding of cancer cells, endothelial-tumor cell interactions govern extravasation to the bone and potentially represent one of the first points of action for antimetastatic treatment. The ephrin-B2-EphB4 pathway controls cellular interactions by inducing repulsive or adhesive properties, depending on forward or reverse signaling. Here, we report that in an in vivo metastatic melanoma model, ephrin-B2-mediated activation of EphB4 induces tumor cell repulsion from bone endothelium, translating in reduced spinal bone metastatic loci and improved neurological function. Selective ephrin-B2 depletion in endothelial cells or EphB4 inhibition increases bone metastasis and shortens the time window to hind-limb locomotion deficit from spinal cord compression. EphB4 overexpression in melanoma cells ameliorates the metastatic phenotype and improves neurological outcome. Timely harvesting of bone tissue after tumor cell injection and intravital bone microscopy revealed less tumor cells attached to ephrin-B2-positive endothelial cells. These results suggest that ephrin-B2-EphB4 communication influences bone metastasis formation by altering melanoma cell repulsion/adhesion to bone endothelial cells, and represents a molecular target for therapeutic intervention.
Collapse
Affiliation(s)
- Thomas Broggini
- Department of Neurosurgery, Universitätsmedizin Charite, D-10117, Berlin, Germany.,Department of Physics, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Andras Piffko
- Department of Neurosurgery, Universitätsmedizin Charite, D-10117, Berlin, Germany.,Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Christian J Hoffmann
- Department of Experimental Neurology, Center for Stroke Research Berlin, Universitätsmedizin Charite, D-10117, Berlin, Germany
| | - Adnan Ghori
- Department of Neurosurgery, Universitätsmedizin Charite, D-10117, Berlin, Germany
| | - Christoph Harms
- Department of Experimental Neurology, Center for Stroke Research Berlin, Universitätsmedizin Charite, D-10117, Berlin, Germany
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Universitätsmedizin Charite, D-10117, Berlin, Germany
| | - Marcus Czabanka
- Department of Neurosurgery, Universitätsmedizin Charite, D-10117, Berlin, Germany.
| |
Collapse
|
17
|
Vieira WF, Malange KF, de Magalhães SF, dos Santos GG, de Oliveira ALR, da Cruz-Höfling MA, Parada CA. Gait analysis correlates mechanical hyperalgesia in a model of streptozotocin-induced diabetic neuropathy: A CatWalk dynamic motor function study. Neurosci Lett 2020; 736:135253. [DOI: 10.1016/j.neulet.2020.135253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/02/2020] [Accepted: 07/14/2020] [Indexed: 01/03/2023]
|
18
|
Preventive hypothermia as a neuroprotective strategy for paclitaxel-induced peripheral neuropathy. Pain 2020; 160:1505-1521. [PMID: 30839425 DOI: 10.1097/j.pain.0000000000001547] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a severe adverse effect that occurs secondary to anticancer treatments and has no known preventive or therapeutic strategy. Therapeutic hypothermia has been shown to be effective in protecting against central and peripheral nervous system injuries. However, the effects of therapeutic hypothermia on CIPN have rarely been explored. We induced lower back hypothermia (LBH) in an established paclitaxel-induced CIPN rat model and found that the paclitaxel-induced impairments observed in behavioral, electrophysiological, and histological impairments were inhibited by LBH when applied at an optimal setting of 24°C to the sciatic nerve and initiated 90 minutes before paclitaxel infusion. Lower back hypothermia also inhibited the paclitaxel-induced activation of astroglia and microglia in the spinal cord and macrophage infiltration into and neuronal injury in the dorsal root ganglia and sciatic nerves. Furthermore, LBH decreased the local blood flow and local tissue concentrations of paclitaxel. Finally, in NOD/SCID mice inoculated with cancer cells, the antiproliferative effect of paclitaxel was not affected by the distal application of LBH. In conclusion, our findings indicate that early exposure to regional hypothermia alleviates paclitaxel-induced peripheral neuropathy. Therapeutic hypothermia may therefore represent an economical and nonpharmaceutical preventive strategy for CIPN in patients with localized solid tumors.
Collapse
|
19
|
James RE, Schalks R, Browne E, Eleftheriadou I, Munoz CP, Mazarakis ND, Reynolds R. Persistent elevation of intrathecal pro-inflammatory cytokines leads to multiple sclerosis-like cortical demyelination and neurodegeneration. Acta Neuropathol Commun 2020; 8:66. [PMID: 32398070 PMCID: PMC7218553 DOI: 10.1186/s40478-020-00938-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 12/12/2022] Open
Abstract
Analysis of isolated meninges and cerebrospinal fluid (CSF) of post-mortem MS cases has shown increased gene and protein expression for the pro-inflammatory cytokines: tumour necrosis factor (TNF) and interferon-γ (IFNγ). Here we tested the hypothesis that persistent production of these cytokines in the meningeal compartment and diffusion into underlying GM can drive chronic MS-like GM pathology. Lentiviral transfer vectors were injected into the sagittal sulcus of DA rats to deliver continuous expression of TNF + IFNγ transgenes in the meninges and the resulting neuropathology analysed after 1 and 2 months. Injection of TNF + IFNγ viral vectors, with or without prior MOG immunisation, induced extensive immune cell infiltration (CD4+ and CD8+ T-cells, CD79a + B-cells and macrophages) in the meninges by 28 dpi, which remained at 2 months. Control GFP viral vector did not induce infiltration. Subpial demyelination was seen underlying these infiltrates, which was partly dependant on prior myelin oligodendrocyte glycoprotein (MOG) immunisation. A significant decrease in neuronal numbers was seen at 28 and 56 days in cortical layers II-V that was independent of MOG immunisation. RNA analysis at 28 dpi showed an increase in expression of necroptotic pathway genes, including RIP3, MLKL, cIAP2 and Nox2. PhosphoRIP3+ and phosphoMLKL+ neurons were present in TNF + IFNγ vector injected animals, indicating activation of necroptosis. Our results suggest that persistent expression of TNF in the presence of IFNγ is a potent inducer of meningeal inflammation and can activate TNF signalling pathways in cortical cells leading to neuronal death and subpial demyelination and thus may contribute to clinical progression in MS.
Collapse
|
20
|
Hill RZ, Bautista DM. Getting in Touch with Mechanical Pain Mechanisms. Trends Neurosci 2020; 43:311-325. [DOI: 10.1016/j.tins.2020.03.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/14/2020] [Accepted: 03/04/2020] [Indexed: 01/10/2023]
|
21
|
Bruna J, Alberti P, Calls-Cobos A, Caillaud M, Damaj MI, Navarro X. Methods for in vivo studies in rodents of chemotherapy induced peripheral neuropathy. Exp Neurol 2020; 325:113154. [PMID: 31837318 PMCID: PMC7105293 DOI: 10.1016/j.expneurol.2019.113154] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022]
Abstract
Peripheral neuropathy is one of the most common, dose limiting, and long-lasting disabling adverse events of chemotherapy treatment. Unfortunately, no treatment has proven efficacy to prevent this adverse effect in patients or improve the nerve regeneration, once it is established. Experimental models, particularly using rats and mice, are useful to investigate the mechanisms related to axonal or neuronal degeneration and target loss of function induced by neurotoxic drugs, as well as to test new strategies to prevent the development of neuropathy and to improve functional restitution. Therefore, objective and reliable methods should be applied for the assessment of function and innervation in adequately designed in vivo studies of CIPN, taking into account the impact of age, sex and species/strains features. This review gives an overview of the most useful methods to assess sensory, motor and autonomic functions, electrophysiological and morphological tests in rodent models of peripheral neuropathy, focused on CIPN. We include as well a proposal of protocols that may improve the quality and comparability of studies undertaken in different laboratories. It is recommended to apply more than one functional method for each type of function, and to perform parallel morphological studies in the same targets and models.
Collapse
Affiliation(s)
- Jordi Bruna
- Unit of Neuro-Oncology, Hospital Universitari de Bellvitge, Institut Català d'Oncologia L'Hospitalet, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, University Milano Bicocca, Monza, Italy; NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Aina Calls-Cobos
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Martial Caillaud
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.
| |
Collapse
|
22
|
Chine VB, Au NPB, Ma CHE. Therapeutic benefits of maintaining mitochondrial integrity and calcium homeostasis by forced expression of Hsp27 in chemotherapy-induced peripheral neuropathy. Neurobiol Dis 2019; 130:104492. [DOI: 10.1016/j.nbd.2019.104492] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/21/2019] [Accepted: 06/05/2019] [Indexed: 01/24/2023] Open
|
23
|
Peng Q, Mechanic J, Shoieb A, Pardo ID, Schaevitz L, Fenyk-Melody J, Vitsky A, Boucher M, Somps C, Cook JC, Liu CN. Circulating microRNA and automated motion analysis as novel methods of assessing chemotherapy-induced peripheral neuropathy in mice. PLoS One 2019; 14:e0210995. [PMID: 30677061 PMCID: PMC6345499 DOI: 10.1371/journal.pone.0210995] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 01/02/2019] [Indexed: 11/18/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CiPN) is a serious adverse effect in the clinic, but nonclinical assessment methods in animal studies are limited to labor intensive behavioral tests or semi-quantitative microscopic evaluation. Hence, microRNA (miRNA) biomarkers and automated in-life behavioral tracking were assessed for their utility as non-invasive methods. To address the lack of diagnostic biomarkers, we explored miR-124, miR-183 and miR-338 in a CiPN model induced by paclitaxel, a well-known neurotoxic agent. In addition, conventional and Vium's innovative Digital Vivarium technology-based in-life behavioral tests and postmortem microscopic examination of the dorsal root ganglion (DRG) and the sciatic nerve were performed. Terminal blood was collected on days 8 or 16, after 20 mg/kg paclitaxel was administered every other day for total of 4 or 7 doses, respectively, for plasma miRNA quantification by RT-qPCR. DRG and sciatic nerve samples were collected from mice sacrificed on day 16 for miRNA quantification. Among the three miRNAs analyzed, only miR-124 was statistically significantly increased (5 fold and 10 fold on day 8 and day 16, respectively). The increase in circulating miR-124 correlated with cold allodynia and axonal degeneration in both DRG and sciatic nerve. Automated home cage motion analysis revealed for the first time that nighttime motion was significantly decreased (P < 0.05) in paclitaxel-dosed animals. Although both increase in circulating miR-124 and decrease in nighttime motion are compelling, our results provide positive evidence warranting further testing using additional peripheral nerve toxicants and diverse experimental CiPN models.
Collapse
Affiliation(s)
- Qinghai Peng
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, La Jolla, California, United States of America
| | | | - Ahmed Shoieb
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Groton, Connecticut, United States of America
| | - Ingrid D. Pardo
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Groton, Connecticut, United States of America
| | | | - Judith Fenyk-Melody
- Comparative Medicine, Pfizer Worldwide Research & Development, Cambridge, Massachusetts, United States of America
| | - Allison Vitsky
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, La Jolla, California, United States of America
| | - Magalie Boucher
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Cambridge, Massachusetts, United States of America
| | - Chris Somps
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Groton, Connecticut, United States of America
| | - Jon C. Cook
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Groton, Connecticut, United States of America
| | - Chang-Ning Liu
- Drug Safety Research & Development, Pfizer Worldwide Research & Development, Groton, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
24
|
Xu Y, Tian NX, Bai QY, Chen Q, Sun XH, Wang Y. Gait Assessment of Pain and Analgesics: Comparison of the DigiGait™ and CatWalk™ Gait Imaging Systems. Neurosci Bull 2019; 35:401-418. [PMID: 30659524 PMCID: PMC6527535 DOI: 10.1007/s12264-018-00331-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/13/2018] [Indexed: 12/17/2022] Open
Abstract
Investigation of pain requires measurements of nociceptive sensitivity and other pain-related behaviors. Recent studies have indicated the superiority of gait analysis over traditional evaluations (e.g., skin sensitivity and sciatic function index [SFI]) in detecting subtle improvements and deteriorations in animal models. Here, pain-related gait parameters, whose criteria include (1) alteration in pain models, (2) correlation with nociceptive threshold, and (3) normalization by analgesics, were identified in representative models of neuropathic pain (spared nerve injury: coordination data) and inflammatory pain (intraplantar complete Freund’s adjuvant: both coordination and intensity data) in the DigiGait™ and CatWalk™ systems. DigiGait™ had advantages in fixed speed (controlled by treadmill) and dynamic SFI, while CatWalk™ excelled in intrinsic velocity, intensity data, and high-quality 3D images. Insights into the applicability of each system may provide guidance for selecting the appropriate gait imaging system for different animal models and optimization for future pain research.
Collapse
Affiliation(s)
- Yu Xu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100083, China
| | - Na-Xi Tian
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100083, China
| | - Qing-Yang Bai
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100083, China
| | - Qi Chen
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100083, China
| | - Xiao-Hong Sun
- Department of Neurobiology, Capital Medical University, Beijing, 100069, China
| | - Yun Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100083, China.
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
25
|
|
26
|
Huehnchen P, Boehmerle W, Endres M. High salt diet ameliorates functional, electrophysiological and histological characteristics of murine spontaneous autoimmune polyneuropathy. Neurobiol Dis 2018; 124:240-247. [PMID: 30468863 DOI: 10.1016/j.nbd.2018.11.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/17/2018] [Accepted: 11/19/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND It was previously reported that high salt dietary conditions can drive autoimmunity and worsen severity and symptoms of autoimmune diseases. Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is a common autoimmune condition of the peripheral nervous system which leads to progressive paralysis and sensory deficits due to a demyelination and secondary axonal loss of peripheral nerves. We used a previously described model with a knockout of CD86 in non-obese diabetic mice (CD86-/- NOD), which results in the spontaneous development of an autoimmune peripheral neuropathy similar to CIDP and investigated the influence of a high salt diet on functional impairment, electrophysiological parameters, demyelination and neuroinflammation in these mice. METHODS At seven weeks of age, asymptomatic female CD86-/- NOD mice were randomly assigned to a normal or high salt diet containing 4% sodium chloride in food and 1% in water. The diet was continued for a total of 30 weeks. RESULTS Mice on the high salt diet showed a delayed onset of clinical symptoms and an ameliorated disease course with a reduced decline of locomotor function. Furthermore, electrophysiological parameters of neuropathy and demyelination were attenuated in mice on the high salt diet, which was confirmed with histological analysis. Additionally, we observed a reduced immune cell infiltration of sciatic nerves in mice which had received the high salt diet. CONCLUSIONS We demonstrate beneficial effects of high salt diet regarding disease progression, functional, electrophysiological and histological parameters in a transgenic mouse model of spontaneous autoimmune neuropathy.
Collapse
Affiliation(s)
- Petra Huehnchen
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Cluster of Excellence NeuroCure, Berlin, Germany; Berlin Institute of Health, 10178 Berlin, Germany.
| | - Wolfgang Boehmerle
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Cluster of Excellence NeuroCure, Berlin, Germany; Berlin Institute of Health, 10178 Berlin, Germany
| | - Matthias Endres
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Cluster of Excellence NeuroCure, Berlin, Germany; Berlin Institute of Health, 10178 Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Center for Stroke Resarch Berlin, Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany; DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| |
Collapse
|
27
|
Maghsoudi OH, Vahedipour A, Robertson B, Spence A. Application of Superpixels to Segment Several Landmarks in Running Rodents. PATTERN RECOGNITION AND IMAGE ANALYSIS 2018. [DOI: 10.1134/s1054661818030082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
28
|
Chine VB, Au NPB, Kumar G, Ma CHE. Targeting Axon Integrity to Prevent Chemotherapy-Induced Peripheral Neuropathy. Mol Neurobiol 2018; 56:3244-3259. [DOI: 10.1007/s12035-018-1301-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023]
|
29
|
Boehmerle W, Huehnchen P, Lee SLL, Harms C, Endres M. TRPV4 inhibition prevents paclitaxel-induced neurotoxicity in preclinical models. Exp Neurol 2018; 306:64-75. [DOI: 10.1016/j.expneurol.2018.04.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/17/2018] [Accepted: 04/27/2018] [Indexed: 12/15/2022]
|
30
|
Abstract
Supplemental Digital Content is Available in the Text. Inhibitors of leukocyte elastase inhibit spontaneous and evoked pain behaviors in mouse models of chronic pain of neuropathic, cancer, and diabetic origins. Neuropathic pain is an integral component of several chronic pain conditions and poses a major health problem worldwide. Despite emerging understanding of mechanisms behind neuropathic pain, the available treatment options are still limited in efficacy or associated with side effects, therefore making it necessary to find viable alternatives. In a genetic screen, we recently identified SerpinA3N, a serine protease inhibitor secreted in response to nerve damage by the dorsal root ganglion neurons and we showed that SerpinA3N acts against induction of neuropathic pain by inhibiting the T-cell- and neutrophil-derived protease, leucocyte elastase (LE). In the current study, via detailed in vivo pharmacology combined with analyses of evoked- and spontaneous pain-related behaviors in mice, we report that on systemic delivery, a single dose of 3 independent LE inhibitors can block established nociceptive hypersensitivity in early and late phases in the spared nerve injury model of traumatic neuropathic pain in mice. We further report the strong efficacy of systemic LE inhibitors in reversing ongoing pain in 2 other clinically relevant mouse models—painful diabetic neuropathy and cancer pain. Detailed immunohistochemical analyses on the peripheral tissue samples revealed that both T-Lymphocytes and neutrophils are the sources of LE on peripheral nerve injury, whereas neutrophils are the primary source of LE in diabetic neuropathic conditions. In summary, our results provide compelling evidence for a strong therapeutic potential of generic LE inhibitors for the treatment of neuropathic pain and other chronic pain conditions harboring a neuropathic pain component.
Collapse
|
31
|
Huehnchen P, Boehmerle W, Endres M. Fingolimod therapy is not effective in a mouse model of spontaneous autoimmune peripheral polyneuropathy. Sci Rep 2018; 8:5648. [PMID: 29618748 PMCID: PMC5884804 DOI: 10.1038/s41598-018-23949-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/21/2018] [Indexed: 12/11/2022] Open
Abstract
Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is an autoimmune disorder, which causes progressive sensory and motor deficits and often results in severe disability. Knockout of the co-stimulatory protein CD86 in mice of the non-obese diabetic background (NoD.129S4-Cd86tm1Shr/JbsJ) results in the development of a spontaneous autoimmune peripheral polyneuropathy (SAPP). We used this previously described transgenic model to study the effects of the sphingosine-1-phosphate receptor agonist fingolimod on SAPP symptoms, functional and electrophysiological characteristics. Compared to two control strains, knockout of CD86 in NOD mice (CD86−/− NOD) resulted in progressive paralysis with distinct locomotor deficits due to a severe sensory-motor axonal-demyelinating polyneuropathy as assessed by electrophysiological measurements. We started fingolimod treatment when CD86−/− NOD mice showed signs of unilateral hind limb weakness and continued at a dose of 1 mg/kg/day for eight weeks. We did not observe any beneficial effects of fingolimod regarding disease progression. In addition, fingolimod did not influence the functional outcome of CD86−/− NOD mice compared to vehicle treatment nor any of the electrophysiological characteristics. In summary, we show that fingolimod treatment has no beneficial effects in autoimmune polyneuropathy, which is in line with recent clinical data obtained in CIDP patients.
Collapse
Affiliation(s)
- Petra Huehnchen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Berlin, Germany. .,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Cluster of Excellence NeuroCure, Berlin, Germany. .,Berlin Institute of Health, 10178, Berlin, Germany.
| | - Wolfgang Boehmerle
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Cluster of Excellence NeuroCure, Berlin, Germany
| | - Matthias Endres
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Cluster of Excellence NeuroCure, Berlin, Germany.,Berlin Institute of Health, 10178, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Stroke Research Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| |
Collapse
|
32
|
von der Ahe D, Huehnchen P, Balkaya M, Peruzzaro S, Endres M, Boehmerle W. Suramin-Induced Neurotoxicity: Preclinical Models and Neuroprotective Strategies. Molecules 2018; 23:molecules23020346. [PMID: 29414872 PMCID: PMC6017835 DOI: 10.3390/molecules23020346] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/23/2018] [Accepted: 02/03/2018] [Indexed: 11/16/2022] Open
Abstract
Suramin is a trypan blue analogon originally developed to treat protozoan infections, which was found to have diverse antitumor effects. One of the most severe side effects in clinical trials was the development of a peripheral sensory-motor polyneuropathy. In this study, we aimed to investigate suramin-induced neuropathy with a focus on calcium (Ca2+) homeostasis as a potential pathomechanism. Adult C57Bl/6 mice treated with a single injection of 250 mg/kg bodyweight suramin developed locomotor and sensory deficits, which were confirmed by electrophysiological measurements showing a predominantly sensory axonal-demyelinating polyneuropathy. In a next step, we used cultured dorsal root ganglia neurons (DRGN) as an in vitro cell model to further investigate underlying pathomechanisms. Cell viability of DRGN was significantly decreased after 24-hour suramin treatment with a calculated IC50 of 283 µM. We detected a suramin-induced Ca2+ influx into DRGN from the extracellular space, which could be reduced with the voltage-gated calcium channel (VGCC) inhibitor nimodipine. Co-incubation of suramin and nimodipine partially improved cell viability of DRGN after suramin exposure. In summary, we describe suramin-induced neurotoxic effects on DRGN as well as potentially neuroprotective agents targeting intracellular Ca2+ dyshomeostasis.
Collapse
Affiliation(s)
- David von der Ahe
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Chariteplatz 1, 10117 Berlin, Germany.
| | - Petra Huehnchen
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Chariteplatz 1, 10117 Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Cluster of Excellence NeuroCure, 10117 Berlin, Germany.
- Berlin Institute of Health, Anna-Louisa-Karsch 2, 10178 Berlin, Germany.
| | - Mustafa Balkaya
- Burke-Cornell Medical Research Institute, White Plains, NY 10605, USA.
| | - Sarah Peruzzaro
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA.
| | - Matthias Endres
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Chariteplatz 1, 10117 Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Cluster of Excellence NeuroCure, 10117 Berlin, Germany.
- Berlin Institute of Health, Anna-Louisa-Karsch 2, 10178 Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Stroke Resarch Berlin, 10117 Berlin, Germany.
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10117 Berlin, Germany.
| | - Wolfgang Boehmerle
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Chariteplatz 1, 10117 Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Cluster of Excellence NeuroCure, 10117 Berlin, Germany.
- Berlin Institute of Health, Anna-Louisa-Karsch 2, 10178 Berlin, Germany.
| |
Collapse
|
33
|
Pharmacological validation of voluntary gait and mechanical sensitivity assays associated with inflammatory and neuropathic pain in mice. Neuropharmacology 2017; 130:18-29. [PMID: 29191755 DOI: 10.1016/j.neuropharm.2017.11.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/09/2017] [Accepted: 11/22/2017] [Indexed: 01/01/2023]
Abstract
The urgent need for more effective analgesic treatment options has prompted a re-evaluation of the behavioral tests used to assess pain in pre-clinical research, with an emphasis on inclusion of more voluntary, un-evoked behavioral assessments of pain. In order to validate voluntary gait analysis and a voluntary mechanical conflict-avoidance assay, we tested mouse models of neuropathy (spared nerve injury) and inflammation (complete Freund's adjuvant) alongside reflexive measures of mechanical and thermal hypersensitivity. To establish whether the observed changes in behavioral responses were pain-related, known analgesics (buprenorphine, gabapentin, carprofen) were also administered. Spared nerve injury persistently altered several gait indices, whereas complete Freund's adjuvant caused only transient changes. Furthermore, known analgesics could not reverse these gait changes, despite demonstrating their previously established efficacy in reflexive measures of mechanical and thermal hypersensitivity. In contrast, the mechanical conflict-avoidance assay demonstrated aversion in mice with neuropathy and inflammation-induced hypersensitivity, which could both be reversed by analgesics. We conclude that voluntary gait changes in rodent neuropathic and inflammatory pain models are not necessarily indicative of pain-related adaptations. On the other hand, mechanical conflict-avoidance represents a valid operant assay for quantifying pain-related behaviors in mice that can be reversed by known analgesics.
Collapse
|
34
|
Kang DW, Choi JG, Moon JY, Kang SY, Ryu Y, Park JB, Kim HW. Automated Gait Analysis in Mice with Chronic Constriction Injury. J Vis Exp 2017:56402. [PMID: 29155722 PMCID: PMC5752418 DOI: 10.3791/56402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The von Frey test is a classical method that has been widely used to examine the sensory function of neuropathic pain animals. However, it has some disadvantages such as subjective data and the requirement of a skilled, experienced experimenter. To date, a variety of modifications have improved the von Frey method, but it still has a few limitations. Recent reports have suggested that gait analysis produces more accurate and objective data from the neuropathic animals. This protocol demonstrates how to perform the automated gait analysis to determine the degree of neuropathic pain in mice. After several days of acclimation, the mice were allowed to walk freely on the glass floor to illuminate footprints. Then, quantification of the footprints and gait were performed through video clips with automatic analysis of various walking parameters, such as area of paw print, swing time, angle of paw, etc. The main purpose of this study is to describe the methodology of automated gait analysis and briefly compare it with data from the classical sensory test using von Frey filament.
Collapse
Affiliation(s)
- Dong-Wook Kang
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University
| | - Jae-Gyun Choi
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University
| | - Ji-Young Moon
- KM Fundamental Research Division, Korea Institute of Oriental Medicine (KIOM)
| | - Suk-Yun Kang
- KM Fundamental Research Division, Korea Institute of Oriental Medicine (KIOM)
| | - Yeonhee Ryu
- KM Fundamental Research Division, Korea Institute of Oriental Medicine (KIOM)
| | - Jin Bong Park
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University
| | - Hyun-Woo Kim
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University; Department of Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston;
| |
Collapse
|
35
|
Liu CN, Berryman E, Zakur D, Shoieb AM, Pardo ID, Boucher M, Somps CJ, Bagi CM, Cook JC. A novel endpoint for the assessment of chemotherapy-induced peripheral neuropathy in rodents: biomechanical properties of peripheral nerve. J Appl Toxicol 2017; 38:193-200. [PMID: 28815646 DOI: 10.1002/jat.3513] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 01/09/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CiPN) is a frequent adverse effect in patients and a leading safety consideration in oncology drug development. Although behavioral assessment and microscopic examination of the nerves and dorsal root ganglia can be incorporated into toxicity studies to assess CiPN risk, more sensitive and less labor-intensive endpoints are often lacking. In this study, rats and mice administered vincristine (75 μg kg-1 day-1 , i.p., for 10 days in rats and 100 μg kg-1 day-1 , i.p., for 11 days in mice, respectively) were employed as the CiPN models. Behavioral changes were assessed during the dosing phase. At necropsy, the sural or sciatic nerve was harvested from the rats and mice, respectively, and assessed for mechanical and histopathological endpoints. It was found that the maximal load and the load/extension ratio were significantly decreased in the nerves collected from the animals dosed with vincristine compared with the vehicle-treated animals (P < 0.05). Additionally, the gait analysis revealed that the paw print areas were significantly increased in mice (P < 0.01), but not in rats following vincristine administration. Light microscopic histopathology of the nerves and dorsal root ganglia were unaffected by vincristine administration. We concluded that ex vivo mechanical properties of the nerves is a sensitive endpoint, providing a new method to predict CiPN in rodent. Gait analysis may also be a useful tool in these pre-clinical animal models.
Collapse
Affiliation(s)
- Chang-Ning Liu
- Worldwide Comparative Medicine, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Edwin Berryman
- Worldwide Comparative Medicine, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - David Zakur
- Worldwide Comparative Medicine, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Ahmed M Shoieb
- Drug Safety Research & Development, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Ingrid D Pardo
- Drug Safety Research & Development, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Magalie Boucher
- Drug Safety Research & Development, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139, USA
| | - Chris J Somps
- Drug Safety Research & Development, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Chedo M Bagi
- Worldwide Comparative Medicine, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| | - Jon C Cook
- Drug Safety Research & Development, Pfizer Worldwide R&D, Groton, Connecticut, 06340, USA
| |
Collapse
|
36
|
Huehnchen P, Boehmerle W, Springer A, Freyer D, Endres M. A novel preventive therapy for paclitaxel-induced cognitive deficits: preclinical evidence from C57BL/6 mice. Transl Psychiatry 2017; 7:e1185. [PMID: 28763058 PMCID: PMC5611721 DOI: 10.1038/tp.2017.149] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 05/18/2017] [Accepted: 06/07/2017] [Indexed: 12/11/2022] Open
Abstract
Chemotherapy-induced central nervous system (CNS) neurotoxicity presents an unmet medical need. Patients often report a cognitive decline in temporal correlation to chemotherapy, particularly for hippocampus-dependent verbal and visuo-spatial abilities. We treated adult C57Bl/6 mice with 12 × 20 mg kg-1 paclitaxel (PTX), mimicking clinical conditions of dose-dense chemotherapy, followed by a pulse of bromodesoxyuridine (BrdU) to label dividing cells. In this model, mice developed visuo-spatial memory impairments, and we measured peak PTX concentrations in the hippocampus of 230 nm l-1, which was sevenfold higher compared with the neocortex. Histologic analysis revealed a reduced hippocampal cell proliferation. In vitro, we observed severe toxicity in slowly proliferating neural stem cells (NSC) as well as human neuronal progenitor cells after 2 h exposure to low nanomolar concentrations of PTX. In comparison, mature post-mitotic hippocampal neurons and cell lines of malignant cells were less vulnerable. In PTX-treated NSC, we observed an increase of intracellular calcium levels, as well as an increased activity of calpain- and caspase 3/7, suggesting a calcium-dependent mechanism. This cell death pathway could be specifically inhibited with lithium, but not glycogen synthase kinase 3 inhibitors, which protected NSC in vitro. In vivo, preemptive treatment of mice with lithium prevented PTX-induced memory deficits and abnormal adult hippocampal neurogenesis. In summary, we identified a molecular pathomechanism, which invokes PTX-induced cytotoxicity in NSC independent of cell cycle status. This pathway could be pharmacologically inhibited with lithium without impairing paclitaxel's tubulin-dependent cytostatic mode of action, enabling a potential translational clinical approach.
Collapse
Affiliation(s)
- P Huehnchen
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Berlin, Germany,Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany,Berlin Institute of Health (BIH), Berlin, Germany
| | - W Boehmerle
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Berlin, Germany,Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany,Berlin Institute of Health (BIH), Berlin, Germany,Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Chariteplatz 1, Berlin 10117, Germany. E-mail:
| | - A Springer
- Großgerätezentrum BioSupraMol, Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - D Freyer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Berlin, Germany,Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Stroke Research Berlin, Berlin, Germany
| | - M Endres
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie, Berlin, Germany,Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany,Berlin Institute of Health (BIH), Berlin, Germany,Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Stroke Research Berlin, Berlin, Germany,German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| |
Collapse
|
37
|
Muralidharan A, Park TSW, Mackie JT, Gimenez LGS, Kuo A, Nicholson JR, Corradini L, Smith MT. Establishment and Characterization of a Novel Rat Model of Mechanical Low Back Pain Using Behavioral, Pharmacologic and Histologic Methods. Front Pharmacol 2017; 8:493. [PMID: 28798688 PMCID: PMC5529395 DOI: 10.3389/fphar.2017.00493] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/10/2017] [Indexed: 11/22/2022] Open
Abstract
Chronic low back pain (LBP), the leading cause of disability globally, is notoriously difficult to treat. Most rodent models of LBP mimic lumbar radicular pain rather than mechanical LBP. Here, we describe establishment of a new rat model of mechanical LBP that is devoid of a neuropathic component. Groups of adult male Sprague Dawley rats were anesthetized and their lumbar L4/L5 and L5/L6 intervertebral disks (IVDs) were punctured (0.5 mm outer diameter, 2mm-deep) 5 (LPB-5X), or 10 (LBP-10X) times per disk. Sham-rats underwent similar surgery, but without disk puncture. Baseline noxious pressure hyperalgesia of lumbar axial deep tissues, mechanical allodynia in the hindpaws and gait were assessed prior to surgery and once-weekly until study completion on day 49. The model was also characterized using pharmacologic and histologic methods. Good animal health was maintained for ≥ 49 days post-surgery. For LBP- but not sham-rats, there was temporal development of noxious pressure hyperalgesia in lumbar axial deep tissues at days 14–49 post-surgery. Importantly, there were no between-group differences in von Frey paw withdrawal thresholds or gait parameters until study completion. On day 49, significant histologic changes were observed in the L4/L5 and L5/L6 IVDs for LBP-10X rats, but not sham-rats. In LBP-10X rats, single bolus doses of morphine produced dose-dependent relief of primary and secondary mechanical hyperalgesia in lumbar axial deep tissues at L4/L5 and L1, respectively. In conclusion, our new rat model has considerable potential for providing novel insight on the pathobiology of mechanical LBP and for analgesic efficacy assessment of novel compounds.
Collapse
Affiliation(s)
- Arjun Muralidharan
- Centre for Integrated Preclinical Drug Development, The University of Queensland, BrisbaneQLD, Australia
| | - Thomas S W Park
- Centre for Integrated Preclinical Drug Development, The University of Queensland, BrisbaneQLD, Australia
| | - John T Mackie
- School of Veterinary Science, The University of Queensland, GattonQLD, Australia
| | - Luiz G S Gimenez
- Centre for Integrated Preclinical Drug Development, The University of Queensland, BrisbaneQLD, Australia
| | - Andy Kuo
- Centre for Integrated Preclinical Drug Development, The University of Queensland, BrisbaneQLD, Australia
| | | | | | - Maree T Smith
- Centre for Integrated Preclinical Drug Development, The University of Queensland, BrisbaneQLD, Australia.,School of Pharmacy, The University of Queensland, BrisbaneQLD, Australia
| |
Collapse
|
38
|
Cao Y, Sun N, Yang JW, Zheng Y, Zhu W, Zhang ZH, Wang XR, Shi GX, Liu CZ. Does acupuncture ameliorate motor impairment after stroke? An assessment using the CatWalk gait system. Neurochem Int 2017; 107:198-203. [DOI: 10.1016/j.neuint.2016.10.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/26/2016] [Accepted: 10/27/2016] [Indexed: 10/20/2022]
|
39
|
McMackin MZ, Henderson CK, Cortopassi GA. Neurobehavioral deficits in the KIKO mouse model of Friedreich's ataxia. Behav Brain Res 2017; 316:183-188. [PMID: 27575947 PMCID: PMC5051948 DOI: 10.1016/j.bbr.2016.08.053] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/24/2016] [Accepted: 08/25/2016] [Indexed: 11/30/2022]
Abstract
Friedreich's Ataxia (FA) is a pediatric neurodegenerative disease whose clinical presentation includes ataxia, muscle weakness, and peripheral sensory neuropathy. The KIKO mouse is an animal model of FA with frataxin deficiency first described in 2002, but neurobehavioral deficits have never been described in this model. The identification of robust neurobehavioral deficits in KIKO mice could support the testing of drugs for FA, which currently has no approved therapy. We tested 13 neurobehavioral tasks to identify a robust KIKO phenotype: Open Field, Grip Strength Test(s), Cylinder, Skilled Forelimb Grasp Task(s), Treadmill Endurance, Locotronic Motor Coordination, Inverted Screen, Treadscan, and Von Frey. Of these, Inverted Screen, Treadscan and Von Frey produced significant neurobehavioral deficits at >8 months of age, and relate to the clinically relevant endpoints of muscle strength and endurance, gait ataxia, and peripheral insensitivity. Thus we identify robust phenotypic measures related to Friedreich's ataxia clinical endpoints which could be used to test effectiveness of potential drug therapy.
Collapse
Affiliation(s)
- Marissa Z McMackin
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA
| | - Chelsea K Henderson
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA
| | - Gino A Cortopassi
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA.
| |
Collapse
|
40
|
Pitzer C, Kuner R, Tappe-Theodor A. EXPRESS: Voluntary and evoked behavioral correlates in neuropathic pain states under different housing conditions. Mol Pain 2016; 12:12/0/1744806916656635. [PMID: 27306409 PMCID: PMC4956152 DOI: 10.1177/1744806916656635] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background There is an urgent need to develop and incorporate novel behavioral tests in classically used preclinical pain models. Most rodent studies are based upon stimulus-evoked hindpaw measurements even though chronic pain is usually a day and night experience. Chronic pain is indeed a debilitating condition that influences the sociability and the ability for voluntary tasks, but the relevant behavioral readouts for these aspects are mostly under-represented in the literature. Moreover, we lack standardization in most behavioral paradigms to guarantee reproducibility and ensure adequate discussion between different studies. This concerns not only the combination, application, and duration of particular behavioral tasks but also the effects of different housing conditions implicating social isolation. Results Our aim was to thoroughly characterize the classically used spared nerve injury model for 12 weeks following surgery. We used a portfolio of classical stimulus-evoked response measurements, detailed gait analysis with two different measuring systems (Dynamic weight bearing (DWB) system and CatWalk), as well as observer-independent voluntary wheel running and home cage monitoring (Laboras system). Additionally, we analyzed the effects of social isolation in all behavioral tasks. We found that evoked hypersensitivity temporally matched changes in static gait parameters, whereas some dynamic gait parameters were changed in a time-dependent manner. Interestingly, voluntary wheel running behavior was not affected in spared nerve injury mice but by social isolation. Besides a reduced climbing activity, spared nerve injury mice did not showed tremendous alterations in the home cage activity. Conclusion This is the first longitudinal study providing detailed insights into various voluntary behavioral parameters related to pain and highlights the importance of social environment on spontaneous non-evoked behaviors in a mouse model of chronic neuropathy. Our results provide fundamental considerations for future experimental planning and discussion of pain-related behavioral changes.
Collapse
|
41
|
Hopkins HL, Duggett NA, Flatters SJ. Chemotherapy-induced painful neuropathy: pain-like behaviours in rodent models and their response to commonly used analgesics. Curr Opin Support Palliat Care 2016; 10:119-128. [PMID: 27054288 PMCID: PMC4982532 DOI: 10.1097/spc.0000000000000204] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW Chemotherapy-induced painful neuropathy (CIPN) is a major dose-limiting side-effect of several widely used chemotherapeutics. Rodent models of CIPN have been developed using a range of dosing regimens to reproduce pain-like behaviours akin to patient-reported symptoms. This review aims to connect recent evidence-based suggestions for clinical treatment to preclinical data. RECENT FINDINGS We will discuss CIPN models evoked by systemic administration of taxanes (paclitaxel and docetaxel), platinum-based agents (oxaliplatin and cisplatin), and the proteasome-inhibitor - bortezomib. We present an overview of dosing regimens to produce CIPN models and their phenotype of pain-like behaviours. In addition, we will discuss how potential, clinically available treatments affect pain-like behaviours in these rodent models, relating those effects to clinical trial data wherever possible. We have focussed on antidepressants, opioids, and gabapentinoids given their broad usage. SUMMARY The review outlines the latest description of the most-relevant rodent models of CIPN enabling comparison between chemotherapeutics, dosing regimen, rodent strain, and sex. Preclinical data support many of the recent suggestions for clinical management of established CIPN and provides evidence for potential treatments warranting clinical investigation. Continued research using rodent CIPN models will provide much needed understanding of the causal mechanisms of CIPN, leading to new treatments for this major clinical problem.
Collapse
Affiliation(s)
- Holly L. Hopkins
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE1 1UL, UK
| | - Natalie A. Duggett
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE1 1UL, UK
| | - Sarah J.L. Flatters
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE1 1UL, UK
| |
Collapse
|
42
|
Wang H, Liu J, Gao G, Wu X, Wang X, Yang H. Protection effect of piperine and piperlonguminine from Piper longum L. alkaloids against rotenone-induced neuronal injury. Brain Res 2016; 1639:214-27. [PMID: 26232071 DOI: 10.1016/j.brainres.2015.07.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 07/18/2015] [Accepted: 07/20/2015] [Indexed: 12/22/2022]
Abstract
Currently available treatment approaches for Parkinson׳s disease (PD) are limited in terms of variety and efficacy. Piper longum L. (PLL; Piperaceae) is used in traditional medicine in Asia and the Pacific Islands, with demonstrated anti-inflammatory and antioxidant activities in preclinical studies, and alkaloid extracts of PLL have shown protective effects in PD models. The present study investigated the mechanistic basis for the observed protective effects of PLL. Rats treated with PLL-derived alkaloids showed improvement in rotenone-induced motor deficits, while reactive oxygen species (ROS) production was decreased, mitochondrial membrane potential was stabilized, and the opening of the mitochondrial permeability transition pore (mPTP)-which is involved in ROS production-was inhibited. In addition, rotenone-induced apoptosis was abrogated in the presence of these alkaloids, while a pretreatment stimulated autophagy, likely mitigating neuronal injury by the removal of damaged mitochondria. These findings provide novel insight into the neuroprotective function of PLL as well as evidence in favor of its use in PD treatment. This article is part of a Special Issue entitled SI: Neuroprotection.
Collapse
Affiliation(s)
- Hao Wang
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Jia Liu
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Ge Gao
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Xia Wu
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Xiaomin Wang
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Hui Yang
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China.
| |
Collapse
|
43
|
Thompson SM, Josey M, Holmes A, Brigman JL. Conditional loss of GluN2B in cortex and hippocampus impairs attentional set formation. Behav Neurosci 2015; 129:105-12. [PMID: 25798630 DOI: 10.1037/bne0000045] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The ability to attend to appropriate stimuli, to plan actions and then alter those actions when environmental conditions change, is essential for an organism to thrive. There is increasing evidence that these executive control processes are mediated in part by N-methyl-D-aspartate receptors (NMDAR). NMDAR subunits confer different physiological properties to the receptor, interact with distinct intracellular postsynaptic scaffolding and signaling molecules and are differentially expressed during development. Recent findings have suggested that the GluN2B subunit may play a unique role in both the acquisition of adaptive choice and the behavioral flexibility required to shift between choices. Here we investigated the role of GluN2B containing NMDARs in the ability to learn, reverse and shift between stimulus dimensions. Mutant mice (floxed-GluN2B x CaMKII-Cre) lacking GluN2B in the dorsal CA1 of the hippocampus and throughout the cortex were tested on an attentional set-shifting task. To explore the role that alterations in motor behavior may have on these behaviors, gross and fine motor behaviors were analyzed in mutant and floxed-control mice. Results show that corticohippocampal loss of GluN2B selectively impaired an initial reversal in a stimulus specific manner and impaired the ability of mutant mice to form an attentional set. Further, GluN2B mice showed normal motor behavior in both overall movement and individual limb behaviors. Together, these results further support the role of NMDAR, and GluN2B in particular, in aspects of executive control including behavioral flexibility and attentional processes.
Collapse
Affiliation(s)
- Shannon M Thompson
- Department of Neurosciences, University of New Mexico School of Medicine
| | - Megan Josey
- Department of Neurosciences, University of New Mexico School of Medicine
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institutes of Health
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico School of Medicine
| |
Collapse
|
44
|
Kobayashi T, Yamauchi K, Matsuura Y, Kuniyoshi K, Takahashi K, Ohtori S. The Effects of Generally Administered Anti-Nerve Growth Factor Receptor (p75NTR) Antibody on Pain-Related Behavior, Dorsal Root Ganglia, and Spinal Glia Activation in a Rat Model of Brachial Plexus Avulsion. J Hand Surg Am 2015; 40:2017-25. [PMID: 26321458 DOI: 10.1016/j.jhsa.2015.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 06/12/2015] [Accepted: 06/12/2015] [Indexed: 02/02/2023]
Abstract
PURPOSE To investigate the effect of intraperitoneal administration of an anti-p75 neurotrophin receptor (p75NTR) antibody on reducing neuropathic pain in a rat model of brachial plexus avulsion (BPA). METHODS We randomly assigned 40 male Wistar rats to 4 groups. In the BPA group, the C8-T1 roots were avulsed from the spinal cord at the lower trunk level, and saline was administered intraperitoneally. In the anti-p75NTR groups, 1 μL or 50 μL anti-p75NTR antibody was administered intraperitoneally after avulsion. In the sham-operated group, the lower trunk level was exposed, and saline was administered intraperitoneally. Mechanical hyperalgesia and pain-induced walking patterns were measured using von Frey filaments and CatWalk gait analysis at various time points until 15 days after administration. At 3 and 15 days after administration, sensory neurons involved in pain perception and satellite glial cells in the ipsilateral C7 dorsal root ganglia were immunolabeled with antibodies against calcitonin gene-related peptide and glial fibrillary acidic protein (GFAP), respectively. At both time points, microglial and astrocyte activation, indicative of spinal pain transmission, were immunohistochemically examined in the ipsilateral dorsal horn of the spinal cord (C7) using anti-ionized calcium-binding adaptor molecule 1 and anti-GFAP antibodies, respectively. RESULTS The gait pattern was significantly improved in both anti-p75NTR groups compared with the BPA group. There were significantly fewer calcitonin gene-related peptide-immunoreactive (IR) neurons, neurons encircled by GFAP-IR satellite glial cells, and GFAP-IR astrocytes in both anti-p75NTR groups compared with the BPA group at both time points. Fewer ionized calcium-binding adaptor molecule 1-IR microglia were quantified in both anti-p75NTR groups compared with the BPA group, but this was only significant at 15 days after administration. CONCLUSIONS Systemic application of the p75NTR inhibitory antibody suppressed neuropathic pain after BPA. CLINICAL RELEVANCE p75NTR may be a potential therapeutic target for the clinical treatment of neuropathic pain in BPA injury.
Collapse
Affiliation(s)
- Tomoko Kobayashi
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Kazuyo Yamauchi
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yusuke Matsuura
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuki Kuniyoshi
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuhisa Takahashi
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Seiji Ohtori
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
45
|
Bober BG, Shah SB. Paclitaxel alters sensory nerve biomechanical properties. J Biomech 2015; 48:3559-67. [PMID: 26321364 DOI: 10.1016/j.jbiomech.2015.07.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/14/2015] [Accepted: 07/18/2015] [Indexed: 11/16/2022]
Abstract
Paclitaxel is an effective chemotherapeutic that, despite its common use, frequently causes debilitating peripheral sensory neuropathy. Paclitaxel binds to and stabilizes microtubules, and through unknown mechanisms, causes abnormal microtubule aggregation. Given that microtubules contribute to the mechanical properties of cells, we tested the hypothesis that paclitaxel treatment would alter the stiffness of sensory nerves. Rat sural nerves were excised and soaked in Ringer's solution with or without paclitaxel. Nerves were secured between a force transducer and actuator, and linearly strained. Stress-strain curves were generated, from which elastic moduli were calculated. Paclitaxel treated nerves exhibited significantly higher moduli in both linear and transition regions of the curve. A composite-tissue model was then generated to estimate the stiffness increase in the cellular fraction of the nerve following paclitaxel treatment. This model was supported experimentally by data on mechanical properties of sural nerves stripped of their epineurium, and area fractions of the cellular and connective tissue components of the rat sural nerve, calculated from immunohistochemical images. Model results revealed that the cellular components of the nerve must stiffen 12x to 115x, depending on the initial axonal modulus assumed, in order to achieve the observed tissue level mechanical changes. Consistent with such an increase, electron microscopy showed increased microtubule aggregation and cytoskeletal packing, suggestive of a more cross-linked cytoskeleton. Overall, our data suggests that paclitaxel treatment induces increased microtubule bundling in axons, which leads to alterations in tissue-level mechanical properties.
Collapse
Affiliation(s)
- Brian G Bober
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Sameer B Shah
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA; Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
46
|
|
47
|
Boehmerle W, Huehnchen P, Peruzzaro S, Balkaya M, Endres M. Electrophysiological, behavioral and histological characterization of paclitaxel, cisplatin, vincristine and bortezomib-induced neuropathy in C57Bl/6 mice. Sci Rep 2014; 4:6370. [PMID: 25231679 PMCID: PMC5377307 DOI: 10.1038/srep06370] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 08/26/2014] [Indexed: 01/12/2023] Open
Abstract
Polyneuropathy is a frequent and potentially severe side effect of clinical tumor chemotherapy. The goal of this study was to characterize paclitaxel-, cisplatin-, vincristine- and bortezomib-induced neuropathy in C57BL/6 mice with a comparative approach. The phenotype of the animals was evaluated at four time points with behavioral and electrophysiological tests, followed by histology. Treatment protocols used in this study were well tolerated and induced a sensory and predominantly axonal polyneuropathy. Behavioral testing revealed normal motor coordination, whereas all mice receiving verum treatment developed mechanical allodynia and distinct gait alterations. Electrophysiological evaluation showed a significant decrease of the caudal sensory nerve action potential amplitude for all cytostatic agents and a moderate reduction of nerve conduction velocity for cisplatin and paclitaxel. This finding was confirmed by histological analysis of the sciatic nerve which showed predominantly axonal damage: Paclitaxel and vincristine affected mostly large myelinated fibers, bortezomib small myelinated fibers and cisplatin damaged all types of myelinated fibers to a similar degree. Neuropathic symptoms developed faster in paclitaxel and vincristine treated animals compared to cisplatin and bortezomib treatment. The animal models in this study can be used to elucidate pathomechanisms underlying chemotherapy-induced polyneuropathy and for the development of novel therapeutic and preventative strategies.
Collapse
Affiliation(s)
- Wolfgang Boehmerle
- 1] Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Berlin, Germany [2] Cluster of Excellence NeuroCure, Charité Universitätsmedizin Berlin, Berlin, Germany [3]
| | - Petra Huehnchen
- 1] Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Berlin, Germany [2] Cluster of Excellence NeuroCure, Charité Universitätsmedizin Berlin, Berlin, Germany [3]
| | - Sarah Peruzzaro
- Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Mustafa Balkaya
- 1] Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Berlin, Germany [2] Center for Stroke Research Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany [3] Neurovascular research laboratory, Department of Radiology, Massachusetts general hospital and Harvard medical school, Boston, MA, USA
| | - Matthias Endres
- 1] Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Berlin, Germany [2] Cluster of Excellence NeuroCure, Charité Universitätsmedizin Berlin, Berlin, Germany [3] Center for Stroke Research Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany [4] German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| |
Collapse
|
48
|
Khan N, Woodruff TM, Smith MT. Establishment and characterization of an optimized mouse model of multiple sclerosis-induced neuropathic pain using behavioral, pharmacologic, histologic and immunohistochemical methods. Pharmacol Biochem Behav 2014; 126:13-27. [PMID: 25223977 DOI: 10.1016/j.pbb.2014.09.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/25/2014] [Accepted: 09/06/2014] [Indexed: 11/19/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) that causes debilitating central neuropathic pain in many patients. Although mouse models of experimental autoimmune encephalomyelitis (EAE) have provided insight on the pathobiology of MS-induced neuropathic pain, concurrent severe motor impairments confound quantitative assessment of pain behaviors over the disease course. To address this issue, we have established and characterized an optimized EAE-mouse model of MS-induced neuropathic pain. Briefly, C57BL/6 mice were immunized with MOG35-55 (200μg) and adjuvants comprising Quil A (45μg) and pertussis toxin (2×250ng). The traditionally used Freund's Complete Adjuvant (FCA) was replaced with Quil A, as FCA itself induces CNS neuroinflammation. Herein, EAE-mice exhibited a mild relapsing-remitting clinical disease course with temporal development of mechanical allodynia in the bilateral hindpaws. Mechanical allodynia was fully developed by 28-30days post-immunization (p.i.) and was maintained until study completion (52-60days p.i.), in the absence of confounding motor deficits. Single bolus doses of amitriptyline (1-7mg/kg), gabapentin (10-50mg/kg) and morphine (0.1-2mg/kg) evoked dose-dependent analgesia in the bilateral hindpaws of EAE-mice; the corresponding ED50s were 1.5, 20 and 1mg/kg respectively. At day 39 p.i. in EAE-mice exhibiting mechanical allodynia in the hindpaws, there was marked demyelination and gliosis in the brain and lumbar spinal cord, mirroring these pathobiologic hallmark features of MS in humans. Our optimized EAE-mouse model of MS-associated neuropathic pain will be invaluable for future investigation of the pathobiology of MS-induced neuropathic pain and for efficacy profiling of novel molecules as potential new analgesics for improved relief of this condition.
Collapse
MESH Headings
- Amines/therapeutic use
- Amitriptyline/therapeutic use
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Brain/pathology
- Cyclohexanecarboxylic Acids/therapeutic use
- Demyelinating Diseases/pathology
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/complications
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Gabapentin
- Gait
- Gliosis/pathology
- Hyperalgesia/chemically induced
- Hyperalgesia/complications
- Hyperalgesia/drug therapy
- Mice
- Morphine/therapeutic use
- Multiple Sclerosis/complications
- Multiple Sclerosis/drug therapy
- Myelin-Oligodendrocyte Glycoprotein
- Neuralgia/complications
- Neuralgia/drug therapy
- Peptide Fragments
- Pertussis Toxin
- Quillaja Saponins
- gamma-Aminobutyric Acid/therapeutic use
Collapse
Affiliation(s)
- Nemat Khan
- The University of Queensland, Center for Integrated Preclinical Drug Development, St Lucia Campus, Brisbane, Queensland 4072, Australia; School of Pharmacy, The University of Queensland, Pharmacy Australia Center of Excellence, Woolloongabba, Brisbane, Queensland 4102, Australia
| | - Trent M Woodruff
- The School of Biomedical Sciences, University of Queensland, St Lucia Campus, Brisbane, Queensland 4072, Australia
| | - Maree T Smith
- The University of Queensland, Center for Integrated Preclinical Drug Development, St Lucia Campus, Brisbane, Queensland 4072, Australia; School of Pharmacy, The University of Queensland, Pharmacy Australia Center of Excellence, Woolloongabba, Brisbane, Queensland 4102, Australia.
| |
Collapse
|
49
|
Specific targeting of neurotoxic side effects and pharmacological profile of the novel cancer stem cell drug salinomycin in mice. J Mol Med (Berl) 2014; 92:889-900. [PMID: 24770997 DOI: 10.1007/s00109-014-1155-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 04/10/2014] [Accepted: 04/11/2014] [Indexed: 12/19/2022]
Abstract
UNLABELLED Salinomycin is a polyether antibiotic which effectively eliminates a variety of cancer stem cells and chemotherapy-resistant tumor cells in vitro and in vivo. One important caveat for its clinical application is the paucity of preclinical pharmacological and safety data. In the present study, we thus aimed to elucidate pharmacokinetic properties of salinomycin and to assess the side effect profile of chronic treatment with this compound in C57Bl/6 mice. In addition, we tested whether neurotoxic side effects can be prevented by interference with the intracellular calcium homeostasis. We observed that salinomycin has a narrow therapeutic index; however, a dose of 5 mg/kg body weight was well tolerated, and analysis of blood parameters as well as organ histology of liver, kidney, skeletal muscle, and heart showed no abnormalities after daily salinomycin injection for 4 weeks. Pharmacokinetic evaluation revealed low micromolar peak concentrations and an almost complete systemic elimination within 5 h after injection. In contrast to low systemic toxicity, typical signs of a sensory polyneuropathy with mechanical and cold allodynia, distinct gait alterations, decreased sensory nerve action potential amplitudes, and loss of myelinated fibers in the sciatic nerve were observed in salinomycin-treated animals. Inhibition of the mitochondrial Na(+)/Ca(2+) exchanger partially prevented the development of salinomycin-induced neuropathy in vivo, an approach which did not reduce salinomycin's antineoplastic efficacy in vitro. Taken together, this study establishes a framework of pharmacokinetic data for future preclinical trials and safety data for translational trials. Furthermore, we established a strategy to reduce salinomycin's off-target neurotoxic effects. KEY MESSAGE Salinomycin has a narrow therapeutic index; a dose of 5 mg/kg is tolerated in mice. Mice treated with salinomycin develop a painful sensory polyneuropathy. An optimized protocol was established to measure salinomycin in serum samples. Inhibition of Na(+)/Ca(2+) exchangers prevents salinomycin-induced neuropathy. Blocking mitochondrial Na(+)/Ca(2+) exchangers does not impair antineoplastic efficacy.
Collapse
|