451
|
Paracha M, Thakar A, Darling RA, Wulff SS, Rule DC, Nair S, Brown TE. Role of cathepsin K in the expression of mechanical hypersensitivity following intra-plantar inflammation. Sci Rep 2022; 12:7108. [PMID: 35501334 PMCID: PMC9061763 DOI: 10.1038/s41598-022-11043-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/14/2022] [Indexed: 11/26/2022] Open
Abstract
Persistent/chronic inflammatory pain involves multiple pathophysiological mechanisms and is far more complex than acute/momentary pain. Current therapeutics for chronic inflammatory pain are often not effective because the etiology responsible for the pain is not addressed by traditional pharmacological treatments. Cathepsin K is a cysteine protease that has mostly been studied in the context of bone and joint disorders. Previous work by others has shown that inhibition of cathepsin K activity reduces osteoarthritis-associated nociception in joints. However, the role of cathepsin K in cutaneous inflammation is understudied. We assessed the effectiveness of genetic deletion or pharmacological inhibition of cathepsin K in male mice on the expression of nocifensive behaviors after formalin injection or mechanical and thermal hypersensitivity after injection of complete Freund’s adjuvant (CFA) into the mouse hind paw. Our data demonstrate that cathepsin K knockout mice (Ctsk−/−) have a reduction in nocifensive behaviors in the formalin test. In addition, Ctsk−/− do not develop mechanical hypersensitivity after CFA injection for up to 7 days. Moreover, we found that inhibition of cathepsin K reduced mechanical hypersensitivity after CFA injection and mRNA levels, protein levels, and cathepsin K activity levels were elevated after CFA injection. Based upon our data, cathepsin K is indicated to play a role in the expression of chemically-induced cutaneous hypersensitivity, as Ctsk−/− mice do not develop mechanical hypersensitivity and show a reduction in nocifensive behaviors. Further research is needed to determine whether attenuating cathepsin K activity may generate a clinically relevant therapeutic.
Collapse
|
452
|
Mayrovitz HN, Maqsood R, Tawakalzada AS. Do Magnetic Fields Have a Place in Treating Vascular Complications in Diabetes? Cureus 2022; 14:e24883. [PMID: 35698680 PMCID: PMC9184174 DOI: 10.7759/cureus.24883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022] Open
Abstract
The use of electromagnetic field therapy (EMFT) is a non-invasive, potential alternative or complementary choice in the treatment of wounds, chronic pain, neuropathy, and other medical conditions, including tissue repair and cell proliferation. Static magnetic fields (SMFs) have been reported to increase microcirculatory blood flow by mediating vasodilation via nitric oxide. Studies report that SMF exposure causes homeostatic, normalizing effects on the vascular tone that may have beneficial effects in situations where tissue perfusion is limited, such as may be present in diabetes. Pulsed electromagnetic fields (PEMFs) have also shown promise in treating diabetic wounds by improving wound healing rates and other attributes. Our purpose was to critically review prior applications of EMFT for relevancy and effectiveness in treating diabetic complications. The goal was to provide information to allow for informed decisions on the possible use of these modalities in the treatment of persons with diabetic complications. The focus was on the following major areas: wound healing, neuropathy, blood glucose control, blood flow, inflammation and oxidative stress.
Collapse
Affiliation(s)
- Harvey N Mayrovitz
- Medical Education, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Davie, USA
| | - Raneem Maqsood
- Medical School, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Aneil S Tawakalzada
- College of Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| |
Collapse
|
453
|
Goder D, Eshkol-Yogev I, Matsliah L, Lemberger M, Harlev M, Furer A, Zilberman M, Egozi D. In vivo study of the efficacy of bupivacaine-eluting novel soy protein wound dressings in a rat burn model. Burns 2022; 48:623-632. [PMID: 34330581 DOI: 10.1016/j.burns.2021.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022]
Abstract
Dealing with wound related pain is an integral part of treatment. Systemic administration of analgesic and anesthetic agents is a common solution for providing pain relief to patients but comes at a risk of severe side effects as well as addiction. To overcome these issues, research efforts were madeto provide a platform for local controlled release of pain killers. We have developed a bilayer soy protein-based wound dressing for the controlled local release of bupivacaine to the wound site. The combination of a dense and a porous layer provides a platform for cell growth and proliferation as well as physical protection to the wound site. The current study focuses on the in vitro bupivacaine release profile from the dressing and the corresponding in vivo results of pain levels in a second-degree burn model on rats. The Rat Grimace Scale method and the Von Frey filaments method were used to quantify both, spontaneous pain and mechanically induced pain. A high burst release of 61.8 ± 1.9% of the loaded drug was obtained during the initial hour, followed by a slower release rate during the following day. The animal trials show that the RGS scores of the bupivacaine-treated group were significantly lower than these of the untreated group, proving a decrease of 51-68% in pain levels during days 1-3 after burn. Hence, successful pain reduction of spontaneous pain as well as mechanically induced pain, for at least three days after burn was achieved. It is concluded that our novel bupivacaine eluting soy protein wound dressings are a promising new concept in the field of local controlled drug release for pain management.
Collapse
Affiliation(s)
- Daniella Goder
- Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv 69978, Israel
| | - Inbar Eshkol-Yogev
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lior Matsliah
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 69978, Israel
| | - Moran Lemberger
- Department of Plastic and Reconstructive Surgery, Kaplan Medical Center, Rehovot, Israel
| | - Mickey Harlev
- Veterinary Service Center, Sackler Faculty of Medicine Tel Aviv University, Tel Aviv 69978, Israel
| | - Ariel Furer
- Medical Corps, Israel Defense Forces, Israel
| | - Meital Zilberman
- Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Dana Egozi
- Department of Plastic and Reconstructive Surgery, Kaplan Medical Center, Rehovot, Israel
| |
Collapse
|
454
|
Alipio JB, Riggs LM, Plank M, Keller A. Environmental Enrichment Mitigates the Long-Lasting Sequelae of Perinatal Fentanyl Exposure in Mice. J Neurosci 2022; 42:3557-3569. [PMID: 35332082 PMCID: PMC9053848 DOI: 10.1523/jneurosci.2083-21.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 11/21/2022] Open
Abstract
The opioid epidemic is a rapidly evolving societal issue driven, in part, by a surge in synthetic opioid use. A rise in fentanyl use among pregnant women has led to a 40-fold increase in the number of perinatally-exposed infants in the past decade. These children are more likely to develop mood-related and somatosensory-related conditions later in life, suggesting that fentanyl may permanently alter neural development. Here, we examined the behavioral and synaptic consequences of perinatal fentanyl exposure in adolescent male and female C57BL/6J mice and assessed the therapeutic potential of environmental enrichment to mitigate these effects. Dams were given ad libitum access to fentanyl (10 µg/ml, per os) across pregnancy and until weaning [postnatal day (PD)21]. Perinatally-exposed adolescent mice displayed hyperactivity (PD45), enhanced sensitivity to anxiogenic environments (PD46), and sensory maladaptation (PD47), sustained behavioral effects that were completely normalized by environmental enrichment (PD21-PD45). Additionally, environmental enrichment normalized the fentanyl-induced changes in the frequency of miniature EPSCs (mEPSCs) of layer 2/3 neurons in the primary somatosensory cortex (S1). We also demonstrate that fentanyl impairs short-term potentiation (STP) and long-term potentiation (LTP) in S1 layer 2/3 neurons, which, instead, exhibit a sustained depression of synaptic transmission that is restored by environmental enrichment. On its own, environmental enrichment suppressed long-term depression (LTD) of control S1 neurons from vehicle-treated mice subjected to standard housing conditions. These results demonstrate that the lasting effects of fentanyl can be ameliorated with a noninvasive intervention introduced during early development.SIGNIFICANCE STATEMENT Illicit use of fentanyl accounts for a large proportion of opioid-related overdose deaths. Children exposed to opioids during development have a higher risk of developing neuropsychiatric disorders later in life. Here, we employ a preclinical model of perinatal fentanyl exposure that recapitulates these long-term impairments and show, for the first time, that environmental enrichment can reverse deficits in somatosensory circuit function and behavior. These findings have the potential to directly inform and guide ongoing efforts to mitigate the consequences of perinatal opioid exposure.
Collapse
Affiliation(s)
- Jason Bondoc Alipio
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Lace Marie Riggs
- Department of Psychiatry, Division of Translational and Basic Science, Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Madeline Plank
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Asaf Keller
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
455
|
Hartrick CT. Exploiting Injury-Induced Peripheral Opioid Receptor Changes in Novel Analgesic Development for Chronic Pain. FRONTIERS IN PAIN RESEARCH 2022; 3:883164. [PMID: 35557854 PMCID: PMC9090307 DOI: 10.3389/fpain.2022.883164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
|
456
|
Medlock L, Sekiguchi K, Hong S, Dura-Bernal S, Lytton WW, Prescott SA. Multiscale Computer Model of the Spinal Dorsal Horn Reveals Changes in Network Processing Associated with Chronic Pain. J Neurosci 2022; 42:3133-3149. [PMID: 35232767 PMCID: PMC8996343 DOI: 10.1523/jneurosci.1199-21.2022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 11/21/2022] Open
Abstract
Pain-related sensory input is processed in the spinal dorsal horn (SDH) before being relayed to the brain. That processing profoundly influences whether stimuli are correctly or incorrectly perceived as painful. Significant advances have been made in identifying the types of excitatory and inhibitory neurons that comprise the SDH, and there is some information about how neuron types are connected, but it remains unclear how the overall circuit processes sensory input or how that processing is disrupted under chronic pain conditions. To explore SDH function, we developed a computational model of the circuit that is tightly constrained by experimental data. Our model comprises conductance-based neuron models that reproduce the characteristic firing patterns of spinal neurons. Excitatory and inhibitory neuron populations, defined by their expression of genetic markers, spiking pattern, or morphology, were synaptically connected according to available qualitative data. Using a genetic algorithm, synaptic weights were tuned to reproduce projection neuron firing rates (model output) based on primary afferent firing rates (model input) across a range of mechanical stimulus intensities. Disparate synaptic weight combinations could produce equivalent circuit function, revealing degeneracy that may underlie heterogeneous responses of different circuits to perturbations or pathologic insults. To validate our model, we verified that it responded to the reduction of inhibition (i.e., disinhibition) and ablation of specific neuron types in a manner consistent with experiments. Thus validated, our model offers a valuable resource for interpreting experimental results and testing hypotheses in silico to plan experiments for examining normal and pathologic SDH circuit function.SIGNIFICANCE STATEMENT We developed a multiscale computer model of the posterior part of spinal cord gray matter (spinal dorsal horn), which is involved in perceiving touch and pain. The model reproduces several experimental observations and makes predictions about how specific types of spinal neurons and synapses influence projection neurons that send information to the brain. Misfiring of these projection neurons can produce anomalous sensations associated with chronic pain. Our computer model will not only assist in planning future experiments, but will also be useful for developing new pharmacotherapy for chronic pain disorders, connecting the effect of drugs acting at the molecular scale with emergent properties of neurons and circuits that shape the pain experience.
Collapse
Affiliation(s)
- Laura Medlock
- Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Kazutaka Sekiguchi
- Drug Developmental Research Laboratory, Shionogi Pharmaceutical Research Center, Toyonaka, Osaka 561-0825, Japan
- State University of New York Downstate Health Science University, Brooklyn, New York 11203
| | - Sungho Hong
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology, Okinawa, 904-0495, Japan
| | - Salvador Dura-Bernal
- State University of New York Downstate Health Science University, Brooklyn, New York 11203
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York 10962
| | - William W Lytton
- State University of New York Downstate Health Science University, Brooklyn, New York 11203
- Kings County Hospital, Brooklyn, New York 11207
| | - Steven A Prescott
- Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
457
|
Narros-Fernández P, Chioua M, Petcu SA, Diez-Iriepa D, Cerrada-Gálvez L, Decouty-Pérez C, Palomino-Antolín A, Ramos E, Farré-Alins V, López-Rodríguez AB, Romero A, Marco-Contelles J, Egea J. Synthesis and Pharmacological Evaluation of New N-Sulfonylureas as NLRP3 Inflammasome Inhibitors: Identification of a Hit Compound to Treat Gout. J Med Chem 2022; 65:6250-6260. [DOI: 10.1021/acs.jmedchem.2c00149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Paloma Narros-Fernández
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Calle Maestro Vives 2, 28009 Madrid, Spain
- IIS-Hospital Universitario de La Princesa, 28006 Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, UAM, Madrid 28029, Spain
| | - Mourad Chioua
- Laboratory of Medicinal Chemistry (IQOG, CSIC), Juan de La Cierva 3, Madrid 28006, Spain
| | - Sonia A. Petcu
- Laboratory of Medicinal Chemistry (IQOG, CSIC), Juan de La Cierva 3, Madrid 28006, Spain
| | - Daniel Diez-Iriepa
- Laboratory of Medicinal Chemistry (IQOG, CSIC), Juan de La Cierva 3, Madrid 28006, Spain
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, Ctra. Madrid-Barcelona km 33.6, Alcalá de Henares 28871, Spain
| | - Laura Cerrada-Gálvez
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Calle Maestro Vives 2, 28009 Madrid, Spain
- IIS-Hospital Universitario de La Princesa, 28006 Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, UAM, Madrid 28029, Spain
| | - Céline Decouty-Pérez
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Calle Maestro Vives 2, 28009 Madrid, Spain
- IIS-Hospital Universitario de La Princesa, 28006 Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, UAM, Madrid 28029, Spain
| | - Alejandra Palomino-Antolín
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Calle Maestro Vives 2, 28009 Madrid, Spain
- IIS-Hospital Universitario de La Princesa, 28006 Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, UAM, Madrid 28029, Spain
| | - Eva Ramos
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, Madrid 28040, Spain
| | - Víctor Farré-Alins
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Calle Maestro Vives 2, 28009 Madrid, Spain
- IIS-Hospital Universitario de La Princesa, 28006 Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, UAM, Madrid 28029, Spain
| | - Ana Belén López-Rodríguez
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Calle Maestro Vives 2, 28009 Madrid, Spain
- IIS-Hospital Universitario de La Princesa, 28006 Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, UAM, Madrid 28029, Spain
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, Madrid 28040, Spain
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC), Juan de La Cierva 3, Madrid 28006, Spain
| | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Calle Maestro Vives 2, 28009 Madrid, Spain
- IIS-Hospital Universitario de La Princesa, 28006 Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, UAM, Madrid 28029, Spain
| |
Collapse
|
458
|
de la Puente B, Zamanillo D, Romero L, Carceller A, Vela JM, Merlos M, Portillo-Salido E. Comprehensive Preclinical Assessment of Sensory, Functional, Motivational-Affective, and Neurochemical Outcomes in Neuropathic Pain: The Case of the Sigma-1 Receptor. ACS Pharmacol Transl Sci 2022; 5:240-254. [PMID: 35434530 PMCID: PMC9003638 DOI: 10.1021/acsptsci.2c00005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Indexed: 12/19/2022]
Abstract
Chronic pain remains a major health problem and is currently facing slow drug innovation. New drug treatments should address not only the sensory-discriminative but also functional and motivational-affective components of chronic pain. In a mouse model of neuropathic pain induced by partial sciatic nerve ligation (PSNL), we analyzed sensory and functional-like outcomes by hindpaw mechanical stimulation and automated gait analysis (CatWalk). We characterized over time a reward-seeking task based on diminished motivation for natural reinforcers (anhedonic-like behavior). To differentiate the appetitive ("wanting") and consummatory ("liking") aspects of motivational behavior, we quantified the latency and number of approaches to eat white chocolate, as well as the eating duration and amount consumed. We explored a putative chronic pain-induced dysregulation of monoamine function by measuring monoamine levels in the nucleus accumbens (NAc), a well-known brain reward area. Finally, we investigated the role of sigma-1 receptor (σ1R) modulation, a nonopioid target, in these multiple dimensions by genetic deletion and pharmacological dose-response studies. After 6 weeks, PSNL increased the approach latency and reduced the consumption of white chocolate in 20-25% of the mice, while around 50-60% had one or the other parameter affected independently. After 10 weeks, sham-operated mice also displayed anhedonic-like behavior. PSNL was associated with reduced extracellular baseline dopamine and increased norepinephrine in the NAc and with a suppression of increased dopamine and serotonin efflux in response to the rewarding stimulus. Genetic and pharmacological blockade of σ1R relieved these multiple alterations in nerve-injured mice. We comprehensively describe sensory, functional, and depression-like impairment of key components of motivated behavior associated with nerve injury. We provide a neurochemical substrate for the depressed mesocorticolimbic reward processing in chronic pain, with a potentially increased translational value. Our results also highlight σ1R for the therapeutic intervention of neuropathic pain.
Collapse
Affiliation(s)
| | - Daniel Zamanillo
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Luz Romero
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Alicia Carceller
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - José Miguel Vela
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Manuel Merlos
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | | |
Collapse
|
459
|
Sok D, Raval S, McKinney J, Drissi H, Mason A, Mautner K, Kaiser JM, Willett NJ. NSAIDs Reduce Therapeutic Efficacy of Mesenchymal Stromal Cell Therapy in a Rodent Model of Posttraumatic Osteoarthritis. Am J Sports Med 2022; 50:1389-1398. [PMID: 35420503 DOI: 10.1177/03635465221083610] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Intra-articular injections of human mesenchymal stromal cells (hMSCs) have shown promise in slowing cartilage degradation in posttraumatic osteoarthritis (PTOA). Clinical use of cell therapies for osteoarthritis has accelerated in recent years without sufficient scientific evidence defining best-use practices. Common recommendations advise patients to avoid nonsteroidal anti-inflammatory drug (NSAID) use before and after cell injection over concerns that NSAIDs may affect therapeutic efficacy. Recommendations to restrict NSAID use are challenging for patients, and it is unclear if patients are compliant. HYPOTHESIS NSAIDs will reduce the efficacy of hMSC therapy in treating a preclinical model of PTOA. STUDY DESIGN Controlled laboratory study. METHODS Lewis rats underwent medial meniscal transection (MMT) surgery to induce PTOA or a sham (sham group) surgery that did not progress to PTOA. Rats received naproxen solution orally daily before (Pre-NSAID group) or after (Post-NSAID group) hMSC treatment, throughout the course of the experiment (Full-NSAID group), or received hMSCs without NSAIDs (No NSAID). Cartilage morphology and composition were quantified using contrast-enhanced micro-computed tomography and histology. Pain (secondary allodynia) was measured using a von Frey filament. RESULTS Injection of hMSCs attenuated cartilage degeneration associated with MMT. hMSCs prevented proteoglycan loss, maintained smooth cartilage surfaces, reduced cartilage lesions, reduced mineralized osteophyte formation, and reduced pain by week 7. The Pre-NSAID group had decreased proteoglycan levels compared with the hMSC group, although there were no other significant differences. Thus, pretreatment with NSAIDs had minimal effects on the therapeutic benefits of hMSC injections. The Post-NSAID and Full-NSAID groups, however, exhibited significantly worse osteoarthritis than the hMSC-only group, with greater proteoglycan loss, surface roughness, osteophyte volume, and pain. CONCLUSION Use of NSAIDs before hMSC injection minimally reduced the therapeutic benefits for PTOA, which included preservation of cartilage surface integrity as well as a reduction in osteophytes. Use of NSAIDs after injections, however, substantially reduced the therapeutic efficacy of cellular treatment. CLINICAL RELEVANCE Our data support the clinical recommendation of avoiding NSAID use after hMSC injection but suggest that using NSAIDs before treatment may not substantially diminish the therapeutic efficacy of cell treatment.
Collapse
Affiliation(s)
- Daniel Sok
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sarvgna Raval
- Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta Veterans Affairs Hospital, Atlanta, Georgia, USA
| | - Jay McKinney
- Emory University School of Medicine, Atlanta, Georgia, USA.,Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Hicham Drissi
- Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta Veterans Affairs Hospital, Atlanta, Georgia, USA
| | - Amadeus Mason
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ken Mautner
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jarred M Kaiser
- Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta Veterans Affairs Hospital, Atlanta, Georgia, USA
| | - Nick J Willett
- Emory University School of Medicine, Atlanta, Georgia, USA.,Georgia Institute of Technology, Atlanta, Georgia, USA.,Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| |
Collapse
|
460
|
Bagues A, López-Tofiño Y, Llorente-Berzal Á, Abalo R. Cannabinoid drugs against chemotherapy-induced adverse effects: focus on nausea/vomiting, peripheral neuropathy and chemofog in animal models. Behav Pharmacol 2022; 33:105-129. [PMID: 35045012 DOI: 10.1097/fbp.0000000000000667] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Although new drugs are being developed for cancer treatment, classical chemotherapeutic agents are still front-line therapies, despite their frequent association with severe side effects that can hamper their use. Cannabinoids may prevent or palliate some of these side effects. The aim of the present study is to review the basic research which has been conducted evaluating the effects of cannabinoid drugs in the treatment of three important side effects induced by classical chemotherapeutic agents: nausea and vomiting, neuropathic pain and cognitive impairment. Several published studies have demonstrated that cannabinoids are useful in preventing and reducing the nausea, vomits and neuropathy induced by different chemotherapy regimens, though other side effects can occur, such as a reduction of gastrointestinal motility, along with psychotropic effects when using centrally-acting cannabinoids. Thus, peripherally-acting cannabinoids and new pharmacological options are being investigated, such as allosteric or biased agonists. Additionally, due to the increase in the survival of cancer patients, there are emerging data that demonstrate an important cognitive deterioration due to chemotherapy, and because the cannabinoid drugs have a neuroprotective effect, they could be useful in preventing chemotherapy-induced cognitive impairment (as demonstrated through studies in other neurological disorders), but this has not yet been tested. Thus, although cannabinoids seem a promising therapeutic approach in the treatment of different side effects induced by chemotherapeutic agents, future research will be necessary to find pharmacological options with a safer profile. Moreover, a new line of research awaits to be opened to elucidate their possible usefulness in preventing cognitive impairment.
Collapse
Affiliation(s)
- Ana Bagues
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón
- High Performance Research Group in Experimental Pharmacology (PHARMAKOM-URJC)
- Unidad Asociada I+D+i del Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Yolanda López-Tofiño
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System NeuGut-URJC
| | - Álvaro Llorente-Berzal
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland
- Centre for Pain Research and Galway Neuroscience Centre, NCBES, National University of Ireland, Galway, Ireland
| | - Raquel Abalo
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón
- Unidad Asociada I+D+i del Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System NeuGut-URJC
- Grupo de Trabajo de Ciencias Básicas en Dolor y Analgesia de la Sociedad Española del Dolor, Madrid, Spain
| |
Collapse
|
461
|
Couly S, Goguadze N, Yasui Y, Kimura Y, Wang SM, Sharikadze N, Wu HE, Su TP. Knocking Out Sigma-1 Receptors Reveals Diverse Health Problems. Cell Mol Neurobiol 2022; 42:597-620. [PMID: 33095392 PMCID: PMC8062587 DOI: 10.1007/s10571-020-00983-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
Sigma-1 receptor (Sig-1R) is a protein present in several organs such as brain, lung, and heart. In a cell, Sig-1R is mainly located across the membranes of the endoplasmic reticulum and more specifically at the mitochondria-associated membranes. Despite numerous studies showing that Sig-1R could be targeted to rescue several cellular mechanisms in different pathological conditions, less is known about its fundamental relevance. In this review, we report results from various studies and focus on the importance of Sig-1R in physiological conditions by comparing Sig-1R KO mice to wild-type mice in order to investigate the fundamental functions of Sig-1R. We note that the Sig-1R deletion induces cognitive, psychiatric, and motor dysfunctions, but also alters metabolism of heart. Finally, taken together, observations from different experiments demonstrate that those dysfunctions are correlated to poor regulation of ER and mitochondria metabolism altered by stress, which could occur with aging.
Collapse
Affiliation(s)
- Simon Couly
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA.
| | - Nino Goguadze
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Yuriko Kimura
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Shao-Ming Wang
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Nino Sharikadze
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Hsiang-En Wu
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| |
Collapse
|
462
|
Face detection and grimace scale prediction of white furred mice. MACHINE LEARNING WITH APPLICATIONS 2022. [DOI: 10.1016/j.mlwa.2022.100312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
463
|
Tappe-Theodor A, Pitzer C, Lewejohann L, Jirkof P, Siegeler K, Segelcke A, Drude N, Pradier B, Pogatzki-Zahn E, Hollinderbäumer B, Segelcke D. The “WWHow” Concept for Prospective Categorization of Post-operative Severity Assessment in Mice and Rats. Front Vet Sci 2022; 9:841431. [PMID: 35372532 PMCID: PMC8964947 DOI: 10.3389/fvets.2022.841431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
The prospective severity assessment in animal experiments in the categories' non-recovery, mild, moderate, and severe is part of each approval process and serves to estimate the harm/benefit. Harms are essential for evaluating ethical justifiability, and on the other hand, they may represent confounders and effect modifiers within an experiment. Catalogs and guidelines provide a way to assess the experimental severity prospectively but are limited in adaptation due to their nature of representing particular examples without clear explanations of the assessment strategies. To provide more flexibility for current and future practices, we developed the modular Where-What-How (WWHow) concept, which applies findings from pre-clinical studies using surgical-induced pain models in mice and rats to provide a prospective severity assessment. The WWHow concept integrates intra-operative characteristics for predicting the maximum expected severity of surgical procedures. The assessed severity categorization is mainly congruent with examples in established catalogs; however, because the WWHow concept is based on anatomical location, detailed analysis of the tissue trauma and other intra-operative characteristics, it enables refinement actions, provides the basis for a fact-based dialogue with authority officials and other stakeholders, and helps to identify confounder factors of study findings.
Collapse
Affiliation(s)
- Anke Tappe-Theodor
- Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, University of Heidelberg, Heidelberg, Germany
| | - Lars Lewejohann
- Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Berlin, Germany
- German Federal Institute for Risk Assessment (BfR), German Center for the Protection of Laboratory Animals (Bf3R), Berlin, Germany
| | - Paulin Jirkof
- Office for Animal Welfare and 3Rs, University of Zurich, Zurich, Switzerland
| | - Katja Siegeler
- Department of Work and Environmental Protection, Westphalian Wilhelms University Muenster, Münster, Germany
| | | | - Natascha Drude
- Berlin Institute of Health (BIH) at Charité, QUEST Center for Responsible Research, Berlin, Germany
| | - Bruno Pradier
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Münster, Germany
| | - Esther Pogatzki-Zahn
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Münster, Germany
| | | | - Daniel Segelcke
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Münster, Germany
- *Correspondence: Daniel Segelcke
| |
Collapse
|
464
|
Blomqvist KJ, Skogster MOB, Kurkela MJ, Rosenholm MP, Ahlström FHG, Airavaara MT, Backman JT, Rauhala PV, Kalso EA, Lilius TO. Systemic hypertonic saline enhances glymphatic spinal cord delivery of lumbar intrathecal morphine. J Control Release 2022; 344:214-224. [PMID: 35301056 DOI: 10.1016/j.jconrel.2022.03.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/13/2022]
Abstract
The blood-brain barrier significantly limits effective drug delivery to central nervous system (CNS) targets. The recently characterized glymphatic system offers a perivascular highway for intrathecally (i.t.) administered drugs to reach deep brain structures. Although periarterial cerebrospinal fluid (CSF) influx and concomitant brain drug delivery can be enhanced by pharmacological or hyperosmotic interventions, their effects on drug delivery to the spinal cord, an important target for many drugs, have not been addressed. Hence, we studied in rats whether enhancement of periarterial flow by systemic hypertonic solution might be utilized to enhance spinal delivery and efficacy of i.t. morphine. We also studied whether the hyperosmolar intervention affects brain or cerebrospinal fluid drug concentrations after systemic administration. Periarterial CSF influx was enhanced by intraperitoneal injection of hypertonic saline (HTS, 5.8%, 20 ml/kg, 40 mOsm/kg). The antinociceptive effects of morphine were characterized, using tail flick, hot plate and paw pressure tests. Drug concentrations in serum, tissue and microdialysis samples were determined by liquid chromatography-tandem mass spectrometry. Compared with isotonic solution, HTS increased concentrations of spinal i.t. administered morphine by 240% at the administration level (T13-L1) at 60 min and increased the antinociceptive effect of morphine in tail flick, hot plate, and paw pressure tests. HTS also independently increased hot plate and paw pressure latencies but had no effect in the tail flick test. HTS transiently increased the penetration of intravenous morphine into the lateral ventricle, but not into the hippocampus. In conclusion, acute systemic hyperosmolality is a promising intervention for enhanced spinal delivery of i.t. administered morphine. The relevance of this intervention should be expanded to other i.t. drugs and brought to clinical trials.
Collapse
Affiliation(s)
- Kim J Blomqvist
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Moritz O B Skogster
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mika J Kurkela
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Marko P Rosenholm
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Fredrik H G Ahlström
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko T Airavaara
- Faculty of Pharmacy and Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Janne T Backman
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pekka V Rauhala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eija A Kalso
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Anaesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital and University of Helsinki, Finland; SleepWell Research Programme, Faculty of Medicine, University of Helsinki, Finland
| | - Tuomas O Lilius
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Emergency Medicine and Services, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
465
|
Antioxidant and Antiaging Properties of Agar Obtained from Brown Seaweed Laminaria digitata (Hudson) in D-Galactose-Induced Swiss Albino Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7736378. [PMID: 35251211 PMCID: PMC8894001 DOI: 10.1155/2022/7736378] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/24/2021] [Accepted: 02/01/2022] [Indexed: 02/02/2023]
Abstract
The present paper explores the antioxidant and antiaging properties of agar extracted from Laminaria digitata (L. digitata) on a D-galactose (D-Gal)-induced mouse model. Experimental mice were divided into four groups: group I comprised of control nontreated mice, group II comprised of D-Gal-induced mice, group III mice were treated with extracted agar after D-Gal induction, and group IV mice were given ascorbic acid as a positive control. Antioxidant enzymes and aging marker proteins declined significantly in group II, whereas they were normal in group III and group IV mice. Expressions of interleukin-1β (IL-1β) in D-Gal-induced mice were significantly enhanced in the liver and brain of the experimental mice, which were otherwise normal in agar-treated mice. Also, IL-6 levels were significantly increased in the liver and reversed in the brain of D-gal mice, while it was regularly in the agar-treated mice. The histopathological analysis of D-Gal-induced mice showed spongiosis and tangles in brain cells, increased fat and decreased collagen contents in the skin, and few dilated sinuses in the hepatic cells. The changes were under control in group III and group IV mice, suggesting the protective effects of agar extracted from L. digitata and ascorbic acid.
Collapse
|
466
|
Ma Q. A functional subdivision within the somatosensory system and its implications for pain research. Neuron 2022; 110:749-769. [PMID: 35016037 PMCID: PMC8897275 DOI: 10.1016/j.neuron.2021.12.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/07/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022]
Abstract
Somatosensory afferents are traditionally classified by soma size, myelination, and their response specificity to external and internal stimuli. Here, we propose the functional subdivision of the nociceptive somatosensory system into two branches. The exteroceptive branch detects external threats and drives reflexive-defensive reactions to prevent or limit injury. The interoceptive branch senses the disruption of body integrity, produces tonic pain with strong aversive emotional components, and drives self-caring responses toward to the injured region to reduce suffering. The central thesis behind this functional subdivision comes from a reflection on the dilemma faced by the pain research field, namely, the use of reflexive-defensive behaviors as surrogate assays for interoceptive tonic pain. The interpretation of these assays is now being challenged by the discovery of distinct but interwoven circuits that drive exteroceptive versus interoceptive types of behaviors, with the conflation of these two components contributing partially to the poor translation of therapies from preclinical studies.
Collapse
Affiliation(s)
- Qiufu Ma
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
467
|
Antinociceptive effect of N-(3-(phenylselanyl)prop-2-yn-1-yl)benzamide in mice: Involvement of 5-HT1A and 5-HT2A/2C receptors. Chem Biol Interact 2022; 359:109918. [DOI: 10.1016/j.cbi.2022.109918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/16/2022] [Accepted: 03/23/2022] [Indexed: 11/23/2022]
|
468
|
Zaki S, Blaker CL, Little CB. OA foundations - experimental models of osteoarthritis. Osteoarthritis Cartilage 2022; 30:357-380. [PMID: 34536528 DOI: 10.1016/j.joca.2021.03.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is increasingly recognised as a disease of diverse phenotypes with variable clinical presentation, progression, and response to therapeutic intervention. This same diversity is readily apparent in the many animal models of OA. However, model selection, study design, and interpretation of resultant findings, are not routinely done in the context of the target human (or veterinary) patient OA sub-population or phenotype. This review discusses the selection and use of animal models of OA in discovery and therapeutic-development research. Beyond evaluation of the different animal models on offer, this review suggests focussing the approach to OA-animal model selection on study objective(s), alignment of available models with OA-patient sub-types, and the resources available to achieve valid and translatable results. How this approach impacts model selection is discussed and an experimental design checklist for selecting the optimal model(s) is proposed. This approach should act as a guide to new researchers and a reminder to those already in the field, as to issues that need to be considered before embarking on in vivo pre-clinical research. The ultimate purpose of using an OA animal model is to provide the best possible evidence if, how, when and where a molecule, pathway, cell or process is important in clinical disease. By definition this requires both model and study outcomes to align with and be predictive of outcomes in patients. Keeping this at the forefront of research using pre-clinical OA models, will go a long way to improving the quality of evidence and its translational value.
Collapse
Affiliation(s)
- S Zaki
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia; Raymond Purves Bone and Joint Research Laboratory, Australia.
| | - C L Blaker
- Raymond Purves Bone and Joint Research Laboratory, Australia; Murray Maxwell Biomechanics Laboratory, The Kolling Institute, University of Sydney Faculty of Medicine and Health, At Royal North Shore Hospital, Australia.
| | - C B Little
- Raymond Purves Bone and Joint Research Laboratory, Australia.
| |
Collapse
|
469
|
Hossain MJ, Kendig MD, Letton ME, Morris MJ, Arnold R. Peripheral Neuropathy Phenotyping in Rat Models of Type 2 Diabetes Mellitus: Evaluating Uptake of the Neurodiab Guidelines and Identifying Future Directions. Diabetes Metab J 2022; 46:198-221. [PMID: 35385634 PMCID: PMC8987683 DOI: 10.4093/dmj.2021.0347] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/25/2022] [Indexed: 11/08/2022] Open
Abstract
Diabetic peripheral neuropathy (DPN) affects over half of type 2 diabetes mellitus (T2DM) patients, with an urgent need for effective pharmacotherapies. While many rat and mouse models of T2DM exist, the phenotyping of DPN has been challenging with inconsistencies across laboratories. To better characterize DPN in rodents, a consensus guideline was published in 2014 to accelerate the translation of preclinical findings. Here we review DPN phenotyping in rat models of T2DM against the 'Neurodiab' criteria to identify uptake of the guidelines and discuss how DPN phenotypes differ between models and according to diabetes duration and sex. A search of PubMed, Scopus and Web of Science databases identified 125 studies, categorised as either diet and/or chemically induced models or transgenic/spontaneous models of T2DM. The use of diet and chemically induced T2DM models has exceeded that of transgenic models in recent years, and the introduction of the Neurodiab guidelines has not appreciably increased the number of studies assessing all key DPN endpoints. Combined high-fat diet and low dose streptozotocin rat models are the most frequently used and well characterised. Overall, we recommend adherence to Neurodiab guidelines for creating better animal models of DPN to accelerate translation and drug development.
Collapse
Affiliation(s)
- Md Jakir Hossain
- Department of Pharmacology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
| | - Michael D. Kendig
- Department of Pharmacology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
| | - Meg E. Letton
- Department of Exercise Physiology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
| | - Margaret J. Morris
- Department of Pharmacology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
| | - Ria Arnold
- Department of Pharmacology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
- Department of Exercise Physiology, School of Medical Sciences, University of New South Wales (UNSW) Sydney, Sydney, Australia
- Department of Exercise and Rehabilitation, School of Medical, Indigenous and Health Science, University of Wollongong, Wollongong, Australia
- Corresponding author: Ria Arnold https://orcid.org/0000-0002-7469-6587 Department of Exercise Physiology, School of Health Sciences, UNSW Sydney, Sydney, NSW 2052, Australia E-mail:
| |
Collapse
|
470
|
Allawadhi P, Khurana A, Sayed N, Godugu C, Vohora D. Ameliorative effect of cerium oxide nanoparticles against Freund's complete adjuvant-induced arthritis. Nanomedicine (Lond) 2022; 17:383-404. [PMID: 35124975 DOI: 10.2217/nnm-2021-0172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 01/11/2022] [Indexed: 12/27/2022] Open
Abstract
Aim: To assess the mechanistic effects of cerium oxide nanoparticles (CONPs) on Freund's complete adjuvant (FCA)-induced rheumatoid arthritis in rats. Methods: CONPs were characterized and evaluated in vitro (RAW 264.7 macrophages) and in vivo (FCA-induced rheumatoid arthritis model). Results:In vitro treatment with CONPs significantly reduced lipopolysaccharide-induced oxidative stress (as evident from dichlorodihydrofluorescein diacetate staining), diminished mitochondrial stress (as observed with tetraethylbenzimidazolylcarbocyanine iodide staining) and reduced superoxide radicals. In vivo, CONPs exhibited anti-rheumatoid arthritis activity, as evident from results of paw volume, x-ray, clinical scoring, levels of cytokines (IL-17, IL-1β, TNF-α and TGF-β1) and histology. Conclusion: We provide preclinical proof that CONPs may be a novel futuristic nanoparticle-based approach for therapy of rheumatoid arthritis.
Collapse
Affiliation(s)
- Prince Allawadhi
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Amit Khurana
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Balanagar, Hyderabad, 500037, Telangana State, India
| | - Nilofer Sayed
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Balanagar, Hyderabad, 500037, Telangana State, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER), Balanagar, Hyderabad, 500037, Telangana State, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| |
Collapse
|
471
|
Ozcan S, Kelestemur MM, Hekim MG, Bulmus O, Bulut F, Bilgin B, Canpolat S, Ozcan M. Asprosin, a novel therapeutic candidate for painful neuropathy: an experimental study in mice. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:325-335. [PMID: 34985531 DOI: 10.1007/s00210-021-02197-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/15/2021] [Indexed: 11/30/2022]
Abstract
Recent studies indicate presence of a strong link between adipokines and neuropathic pain. However, the effects of asprosin, a novel adipokine, on neuropathic pain have not been studied in animal models.Mouse models were employed to investigate the antinociceptive effectiveness of asprosin in the treatment of three types of neuropathic pain, with metabolic (streptozocin/STZ), toxic (oxaliplatin/OXA), and traumatic (sciatic nerve ligation/CCI [chronic constriction nerve injury]) etiologies, respectively. Changes in nociceptive behaviors were assessed relative to controls using thermal (the hot plate and cold plate tests, at 50 °C and 4 °C respectively) and mechanical pain (von Frey test) tests after intraperitoneal (i.p.) administration of asprosin (10 µg/kg) and gabapentin (50 mg/kg) in several times intervals. Besides, possible effect of asprosin on the motor coordination of mice was assessed with a rotarod test. Serum level of asprosin was quantified by ELISA.In neuropathic pain models (STZ, OXA, and CCI), asprosin administration significantly reduced both mechanical and thermal hypersensitivity, indicating that it exhibits a clear-cut antihypersensitivity effect in the analyzed neuropathic pain models. The most effective time of asprosin on pain threshold was observed 60 min after its injection. Also, asprosin displayed no notable effect on the motor activity. Asprosin levels were significantly lower in neuropathic pain compared to healthy group (p < 0.05).The results yielded by the present study suggest that asprosin exhibits an analgesic effect in the neuropathic pain models and may have clinical utility in alleviating chronic pain associated with disease and injury originating from peripheral structures.
Collapse
Affiliation(s)
- Sibel Ozcan
- Department of Anaesthesiology and Reanimation, Faculty of Medicine, Firat University, Elazig, Turkey
| | | | | | - Ozgur Bulmus
- Department of Physiology, Faculty of Medicine, Balikesir University, Balikesir, Turkey
| | - Ferah Bulut
- Department of Biophysics, Faculty of Medicine, Firat University, TR23119, Elazig, Turkey
| | - Batuhan Bilgin
- Department of Biophysics, Faculty of Medicine, Firat University, TR23119, Elazig, Turkey
| | - Sinan Canpolat
- Department of Physiology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Mete Ozcan
- Department of Biophysics, Faculty of Medicine, Firat University, TR23119, Elazig, Turkey.
| |
Collapse
|
472
|
Choi Y, Shin T. Alendronate Enhances Functional Recovery after Spinal Cord Injury. Exp Neurobiol 2022; 31:54-64. [PMID: 35256544 PMCID: PMC8907254 DOI: 10.5607/en21030] [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: 09/09/2021] [Revised: 12/12/2021] [Accepted: 01/12/2022] [Indexed: 11/24/2022] Open
Abstract
Spinal cord injury is a destructive disease characterized by motor/sensory dysfunction and severe inflammation. Alendronate is an anti-inflammatory molecule and may therefore be of benefit in the treatment of the inflammation associated with spinal cord injury. This study aimed to evaluate whether alendronate attenuates motor/sensory dysfunction and the inflammatory response in a thoracic spinal cord clip injury model. Alendronate was intraperitoneally administered at 1 mg/kg/day or 5 mg/kg/day from day (D) 0 to 28 post-injury (PI). The histopathological evaluation showed an alleviation of the inflammatory response, including the infiltration of inflammatory cells, and a decrease in gliosis. Alendronate also led to reductions in the levels of inflammation-related molecules, including mitogen-activated protein kinase, p53, pro-inflammatory cytokines, and pro-inflammatory mediators. Neuro-behavioral assessments, including the Basso, Beattie, and Bresnahan scale for locomotor function, the von Frey filament test, the hot plate test, and the cold stimulation test for sensory function, and the horizontal ladder test for sensorimotor function improved significantly in the alendronate-treated group at D28PI. Taken together, these results suggest that alendronate treatment can inhibit the inflammatory response in spinal cord injury thus improving functional responses.
Collapse
Affiliation(s)
- Yuna Choi
- Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| |
Collapse
|
473
|
Sulima A, Li F, Morgan JB, Truong P, Antoline JFG, Oertel T, Barrientos RC, Torres OB, Beck Z, Imler GH, Deschamps JR, Matyas GR, Jacobson AE, Rice KC. Design, Synthesis, and In Vivo Evaluation of C1-Linked 4,5-Epoxymorphinan Haptens for Heroin Vaccines. Molecules 2022; 27:1553. [PMID: 35268659 PMCID: PMC8911913 DOI: 10.3390/molecules27051553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/16/2022] [Accepted: 02/21/2022] [Indexed: 11/16/2022] Open
Abstract
In our continuing effort to develop effective anti-heroin vaccines as potential medications for the treatment of opioid use disorder, herein we present the design and synthesis of the haptens: 1-AmidoMorHap (1), 1-AmidoMorHap epimer (2), 1 Amido-DihydroMorHap (3), and 1 Amido-DihydroMorHap epimer (4). This is the first report of hydrolytically stable haptenic surrogates of heroin with the attachment site at the C1 position in the 4,5-epoxymorophinan nucleus. We prepared respective tetanus toxoid (TT)-hapten conjugates as heroin vaccine immunogens and evaluated their efficacy in vivo. We showed that all TT-hapten conjugates induced high antibody endpoint titers against the targets but only haptens 2 and 3 can induce protective effects against heroin in vivo. The epimeric analogues of these haptens, 1 and 4, failed to protect mice from the effects of heroin. We also showed that the in vivo efficacy is consistent with the results of the in vitro drug sequestration assay. Attachment of the linker at the C1 position induced antibodies with weak binding to the target drugs. Only TT-2 and TT-3 yielded antibodies that bound heroin and 6-acetyl morphine. None of the TT-hapten conjugates induced antibodies that cross-reacted with morphine, methadone, naloxone, or naltrexone, and only TT-3 interacted weakly with buprenorphine, and that subtle structural difference, especially at the C6 position, can vastly alter the specificity of the induced antibodies. This study is an important contribution in the field of vaccine development against small-molecule targets, providing proof that the chirality at C6 in these epoxymorphinans is a vital key to their effectiveness.
Collapse
Affiliation(s)
- Agnieszka Sulima
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Fuying Li
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Jeffrey Brian Morgan
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Phong Truong
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Joshua F. G. Antoline
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Therese Oertel
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (T.O.); (R.C.B.); (O.B.T.); (Z.B.); (G.R.M.)
| | - Rodell C. Barrientos
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (T.O.); (R.C.B.); (O.B.T.); (Z.B.); (G.R.M.)
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Oscar B. Torres
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (T.O.); (R.C.B.); (O.B.T.); (Z.B.); (G.R.M.)
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Zoltan Beck
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (T.O.); (R.C.B.); (O.B.T.); (Z.B.); (G.R.M.)
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Gregory H. Imler
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA; (G.H.I.); (J.R.D.)
| | - Jeffrey R. Deschamps
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA; (G.H.I.); (J.R.D.)
| | - Gary R. Matyas
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (T.O.); (R.C.B.); (O.B.T.); (Z.B.); (G.R.M.)
| | - Arthur E. Jacobson
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Kenner C. Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| |
Collapse
|
474
|
Muñoz-Lora VRM, Dugonjić Okroša A, Matak I, Del Bel Cury AA, Kalinichev M, Lacković Z. Antinociceptive Actions of Botulinum Toxin A1 on Immunogenic Hypersensitivity in Temporomandibular Joint of Rats. Toxins (Basel) 2022; 14:toxins14030161. [PMID: 35324657 PMCID: PMC8953731 DOI: 10.3390/toxins14030161] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 12/29/2022] Open
Abstract
Botulinum neurotoxin type A1 (BoNT-A) reduces the peripheral peptide and cytokine upregulation in rats with antigen-evoked persistent immunogenic hypersensitivity (PIH) of the temporomandibular joint (TMJ). Herein, we examined the effects of two preparations of BoNT-A, abobotulinumtoxinA (aboBoNT-A; Dysport) and onabotulinumtoxinA (onaBoNT-A; Botox), on spontaneous and evoked nociceptive behaviors, as well as on central neuronal and astroglial activation. The antigen-evoked PIH was induced in rats via repeated systemic and unilateral intra-articular (i.a.) injections of methylated bovine serum albumin (mBSA). Rats were subsequently injected with unilateral i.a. aboBoNT-A (14 U/kg), onaBoNT-A (7 U/kg), or the vehicle (saline). After i.a. treatments, spontaneous and mechanically evoked nocifensive behaviors were assessed before and after the low-dose i.a. formalin (0.5%) challenge. The central effects of BoNT-A were assessed by an immunohistochemical analysis of cleaved synaptosomal-associated protein 25 (cSNAP-25) presence, c-Fos, GFAP, and CGRP expression in the trigeminal nucleus caudalis (TNC). Both BoNT-A preparations similarly reduced the formalin-induced spontaneous pain-related behaviors and mechanical allodynia of the hypernociceptive rats. Likewise, their effects were associated with the central occurrence of cSNAP-25 and reduction of c-Fos and GFAP upregulation in the TNC. BoNT-A antinociceptive activity on the PIH is associated with the toxin axonal transport to trigeminal sensory areas and reduction of neuronal and glial activation in central nociceptive regions.
Collapse
Affiliation(s)
- Victor Ricardo Manuel Muñoz-Lora
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (V.R.M.M.-L.); (I.M.)
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba 13414-903, Brazil;
- Dental Research Division, School of Dentistry, Ibirapuera University, São Paulo 04661-100, Brazil
| | - Ana Dugonjić Okroša
- Department of Pharmacology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia;
| | - Ivica Matak
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (V.R.M.M.-L.); (I.M.)
| | - Altair Antoninha Del Bel Cury
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba 13414-903, Brazil;
| | | | - Zdravko Lacković
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (V.R.M.M.-L.); (I.M.)
- Correspondence: ; Tel.: +385-1-4566-843
| |
Collapse
|
475
|
Salehi MS, Jurek B, Karimi-Haghighi S, Nezhad NJ, Mousavi SM, Hooshmandi E, Safari A, Dianatpour M, Haerteis S, Miyan JA, Pandamooz S, Borhani-Haghighi A. Intranasal application of stem cells and their derivatives as a new hope in the treatment of cerebral hypoxia/ischemia: a review. Rev Neurosci 2022; 33:583-606. [DOI: 10.1515/revneuro-2021-0163] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/13/2022] [Indexed: 02/07/2023]
Abstract
Abstract
Intranasal delivery of stem cells and conditioned medium to target the brain has attracted major interest in the field of regenerative medicine. In pre-clinical investigations during the last ten years, several research groups focused on this strategy to treat cerebral hypoxia/ischemia in neonates as well as adults. In this review, we discuss the curative potential of stem cells, stem cell derivatives, and their delivery route via intranasal application to the hypoxic/ischemic brain. After intranasal application, stem cells migrate from the nasal cavity to the injured area and exert therapeutic effects by reducing brain tissue loss, enhancing endogenous neurogenesis, and modulating cerebral inflammation that leads to functional improvements. However, application of this administration route for delivering stem cells and/or therapeutic substances to the damaged sites requires further optimization to translate the findings of animal experiments to clinical trials.
Collapse
Affiliation(s)
- Mohammad Saied Salehi
- Clinical Neurology Research Center , Shiraz University of Medical Sciences , Shiraz 71936-35899 , Iran
| | - Benjamin Jurek
- Institute of Molecular and Cellular Anatomy , University of Regensburg , Regensburg 93053 , Germany
| | - Saeideh Karimi-Haghighi
- Clinical Neurology Research Center , Shiraz University of Medical Sciences , Shiraz 71936-35899 , Iran
| | - Nahid Jashire Nezhad
- Clinical Neurology Research Center , Shiraz University of Medical Sciences , Shiraz 71936-35899 , Iran
| | - Seyedeh Maryam Mousavi
- Clinical Neurology Research Center , Shiraz University of Medical Sciences , Shiraz 71936-35899 , Iran
| | - Etrat Hooshmandi
- Clinical Neurology Research Center , Shiraz University of Medical Sciences , Shiraz 71936-35899 , Iran
| | - Anahid Safari
- Stem Cells Technology Research Center , Shiraz University of Medical Sciences , Shiraz 71936-35899 , Iran
| | - Mehdi Dianatpour
- Stem Cells Technology Research Center , Shiraz University of Medical Sciences , Shiraz 71936-35899 , Iran
| | - Silke Haerteis
- Institute of Molecular and Cellular Anatomy , University of Regensburg , Regensburg 93053 , Germany
| | - Jaleel A. Miyan
- Faculty of Biology, Medicine & Health, Division of Neuroscience & Experimental Psychology , The University of Manchester , Manchester M13 9PL , UK
| | - Sareh Pandamooz
- Stem Cells Technology Research Center , Shiraz University of Medical Sciences , Shiraz 71936-35899 , Iran
| | - Afshin Borhani-Haghighi
- Clinical Neurology Research Center , Shiraz University of Medical Sciences , Shiraz 71936-35899 , Iran
| |
Collapse
|
476
|
The Surface Amine Group of Ultrasmall Magnetic Iron Oxide Nanoparticles Produce Analgesia in the Spinal Cord and Decrease Long-Term Potentiation. Pharmaceutics 2022; 14:pharmaceutics14020366. [PMID: 35214098 PMCID: PMC8879719 DOI: 10.3390/pharmaceutics14020366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/30/2022] [Accepted: 02/03/2022] [Indexed: 11/19/2022] Open
Abstract
Our previous studies have revealed the ultrasmall superparamagnetic iron oxide in the amine group USPIO-101 has an analgesic effect on inflammatory pain. Here, we further investigated its effect on the spinal cord and brain via electrophysiological and molecular methods. We used a mouse inflammatory pain model, induced by complete Freund’s adjuvant (CFA), and measured pain thresholds via von Frey methods. We also investigated the effects of USPIO-101 via an extracellular electrophysiological recording at the spinal dorsal horn synapses and hippocampal Schaffer collateral-CA1 synapses, respectively. The mRNA expression of pro-inflammatory cytokines was detected by quantitative real-time polymerase chain reaction (RT-qPCR). Our results showed intrathecal USPIO-101 produces similar analgesic behavior in mice with chronic inflammatory pain via intrathecal or intraplantar administration. The potentiated low-frequency stimulation-induced spinal cord long-term potentiation (LTP) at the spinal cord superficial dorsal horn synapses could decrease via USPIO-101 in mice with chronic inflammatory pain. However, the mRNA expression of cyclooxygenase-2 was enhanced with lipopolysaccharide (LPS) stimulation in microglial cells, and we also found USPIO-101 at 30 µg/mL could decrease the magnitude of hippocampal LTP. These findings revealed that intrathecal USPIO-101 presented an analgesia effect at the spinal cord level, but had neurotoxicity risk at higher doses.
Collapse
|
477
|
Antoniazzi C, Belinskaia M, Zurawski T, Kaza SK, Dolly JO, Lawrence GW. Botulinum Neurotoxin Chimeras Suppress Stimulation by Capsaicin of Rat Trigeminal Sensory Neurons In Vivo and In Vitro. Toxins (Basel) 2022; 14:116. [PMID: 35202143 PMCID: PMC8878885 DOI: 10.3390/toxins14020116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/14/2022] [Accepted: 02/01/2022] [Indexed: 02/06/2023] Open
Abstract
Chimeras of botulinum neurotoxin (BoNT) serotype A (/A) combined with /E protease might possess improved analgesic properties relative to either parent, due to inheriting the sensory neurotropism of the former with more extensive disabling of SNAP-25 from the latter. Hence, fusions of /E protease light chain (LC) to whole BoNT/A (LC/E-BoNT/A), and of the LC plus translocation domain (HN) of /E with the neuronal acceptor binding moiety (HC) of /A (BoNT/EA), created previously by gene recombination and expression in E. coli., were used. LC/E-BoNT/A (75 units/kg) injected into the whisker pad of rats seemed devoid of systemic toxicity, as reflected by an absence of weight loss, but inhibited the nocifensive behavior (grooming, freezing, and reduced mobility) induced by activating TRPV1 with capsaicin, injected at various days thereafter. No sex-related differences were observed. c-Fos expression was increased five-fold in the trigeminal nucleus caudalis ipsi-lateral to capsaicin injection, relative to the contra-lateral side and vehicle-treated controls, and this increase was virtually prevented by LC/E-BoNT/A. In vitro, LC/E-BoNT/A or /EA diminished CGRP exocytosis from rat neonate trigeminal ganglionic neurons stimulated with up to 1 µM capsaicin, whereas BoNT/A only substantially reduced the release in response to 0.1 µM or less of the stimulant, in accordance with the /E protease being known to prevent fusion of exocytotic vesicles.
Collapse
Affiliation(s)
| | | | | | | | | | - Gary W. Lawrence
- International Centre for Neurotherapeutics, Dublin City University, Collins Avenue, D09 V209 Dublin, Ireland; (C.A.); (M.B.); (T.Z.); (S.K.K.); (J.O.D.)
| |
Collapse
|
478
|
Andrade-González RD, Perrusquia-Hernández E, Montes-Ángeles CD, Castillo-Díaz LA, Hernández Campos ME, Pérez-Martínez IO. Encoding signs of orofacial neuropathic pain from facial expressions in mice. Arch Oral Biol 2022; 135:105369. [DOI: 10.1016/j.archoralbio.2022.105369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 11/02/2022]
|
479
|
Zhao QX, Wang YH, Wang SC, Xue S, Cao ZX, Sun T. Protectin DX Attenuates Lumbar Radicular Pain of Non-compressive Disc Herniation by Autophagy Flux Stimulation via Adenosine Monophosphate-Activated Protein Kinase Signaling. Front Physiol 2022; 12:784653. [PMID: 35069245 PMCID: PMC8770935 DOI: 10.3389/fphys.2021.784653] [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: 09/28/2021] [Accepted: 12/08/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Neuroinflammation plays a crucial role in initiating and sustaining lumbar radicular pain (LRP). Protectin DX (PDX) has been experimentally verified to possess pro-resolving properties and anti-inflammatory effects. This study aimed to observe the analgesic effects of PDX and its potential mechanisms in LRP rats with non-compressive lumbar disc herniation (NCLDH). Method: Only male rats were selected to avoid gender-related interferences. Rat models of NCLDH were established, and rats were randomly divided into four groups: the sham group, the vehicle group, the PDX (10 ng PDX) group, and the PDX (100 ng PDX) group. Changes in the mechanical withdrawal threshold and thermal withdrawal latency were observed for 7 days. The mRNAs of pro-inflammatory and anti-inflammatory mediators were evaluated via real-time polymerase chain reaction, whereas western blot and immunohistochemistry were separately conducted to assess the expression levels of autophagy-related proteins and adenosine monophosphate-activated protein kinase (AMPK) signaling. Results: Intrathecal delivery of PDX reduced interleukin (IL)-6 and IL-1β mRNA levels and facilitated mRNA transcription of transforming growth factor-β1, with attenuation of mechanical and thermal hyperalgesia in LRP rat models. With the application of nucleus pulposus to the dorsal root ganglion, autophagy flux and AMPK signaling were severely disrupted in the spinal dorsal horns, and intrathecal treatment with PDX could dose-dependently restore the dysfunction of autophagy flux and AMPK signaling. Conclusion: These data suggest that PDX possesses pro-resolving properties and exerts potent analgesic effects in LRP by affecting autophagy flux via AMPK signaling.
Collapse
Affiliation(s)
- Qing-Xiang Zhao
- Department of Pain Management, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pain Management, Binzhou Medical University Hospital, Binzhou, China
| | - Yi-Hao Wang
- Department of Pain Management, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Si-Cong Wang
- Departments of Pain Management, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Song Xue
- Department of Pain Management, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhen-Xin Cao
- Departments of Pain Management, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tao Sun
- Department of Pain Management, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Departments of Pain Management, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
480
|
Xie AX, Iguchi N, Clarkson TC, Malykhina AP. Pharmacogenetic inhibition of lumbosacral sensory neurons alleviates visceral hypersensitivity in a mouse model of chronic pelvic pain. PLoS One 2022; 17:e0262769. [PMID: 35077502 PMCID: PMC8789164 DOI: 10.1371/journal.pone.0262769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
The study investigated the cellular and molecular mechanisms in the peripheral nervous system (PNS) underlying the symptoms of urologic chronic pelvic pain syndrome (UCPPS) in mice. This work also aimed to test the feasibility of reversing peripheral sensitization in vivo in alleviating UCPPS symptoms. Intravesical instillation of vascular endothelial growth factor A (VEGFA) was used to induce UCPPS-like symptoms in mice. Spontaneous voiding spot assays and manual Von Frey tests were used to evaluate the severity of lower urinary tract symptoms (LUTS) and visceral hypersensitivity in VEGFA-instilled mice. Bladder smooth muscle strip contractility recordings (BSMSC) were used to identify the potential changes in myogenic and neurogenic detrusor muscle contractility at the tissue-level. Quantitative real-time PCR (qPCR) and fluorescent immunohistochemistry were performed to compare the expression levels of VEGF receptors and nociceptors in lumbosacral dorsal root ganglia (DRG) between VEGFA-instilled mice and saline-instilled controls. To manipulate primary afferent activity, Gi-coupled Designer Receptors Exclusively Activated by Designer Drugs (Gi-DREADD) were expressed in lumbosacral DRG neurons of TRPV1-Cre-ZGreen mice via targeted adeno-associated viral vector (AAVs) injections. A small molecule agonist of Gi-DREADD, clozapine-N-oxide (CNO), was injected into the peritoneum (i. p.) in awake animals to silence TRPV1 expressing sensory neurons in vivo during physiological and behavioral recordings of bladder function. Intravesical instillation of VEGFA in the urinary bladders increased visceral mechanical sensitivity and enhanced RTX-sensitive detrusor contractility. Sex differences were identified in the baseline detrusor contractility responses and VEGF-induced visceral hypersensitivity. VEGFA instillations in the urinary bladder led to significant increases in the mRNA and protein expression of transient receptor potential cation channel subfamily A member 1 (TRPA1) in lumbosacral DRG, whereas the expression levels of transient receptor potential cation channel subfamily V member 1 (TRPV1) and VEGF receptors (VEGFR1 and VEGFR2) remained unchanged when compared to saline-instilled animals. Importantly, the VEGFA-induced visceral hypersensitivity was reversed by Gi-DREADD-mediated neuronal silencing in lumbosacral sensory neurons. Activation of bladder VEGF signaling causes sensory neural plasticity and visceral hypersensitivity in mice, confirming its role of an UCPPS biomarker as identified by the Multidisciplinary Approach to the Study of Chronic Pelvic Pain (MAPP) research studies. Pharmacogenetic inhibition of lumbosacral sensory neurons in vivo completely reversed VEGFA-induced pelvic hypersensitivity in mice, suggesting the strong therapeutic potential for decreasing primary afferent activity in the treatment of pain severity in UCPPS patients.
Collapse
Affiliation(s)
- Alison Xiaoqiao Xie
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| | - Nao Iguchi
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| | - Taylor C. Clarkson
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| | - Anna P. Malykhina
- Department of Surgery, School of Medicine, Anschutz Medical Campus, University of Colorado, Denver, Colorado, United States of America
| |
Collapse
|
481
|
Karmakar V, Mohammad FS, Baeesa SS, Alexiou A, Sivakumar SR, Ashraf GM. Effect of Cliothosa aurivilli on Paclitaxel-induced Peripheral Neuropathy in Experimental Animals. Mol Neurobiol 2022; 59:2232-2245. [PMID: 35064539 DOI: 10.1007/s12035-021-02685-3] [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: 10/03/2021] [Accepted: 12/08/2021] [Indexed: 11/28/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a serious complication leading to painful episodes of parasthesia and numbness in hands and feet. The present drugs that have been used for symptomatic treatment yield inconclusive results in trials and assorted side effects. Thus, there is a pressing demand for development of therapeutically efficacious strategy to combat CIPN. The present study investigates about the effect of a marine sponge; Cliothosa aurivilli (CA) on paclitaxel (PT)-induced peripheral neuropathy in mice. Peripheral neuropathy was induced by intoxication with chemotherapeutic drug PT (2 mg/kg; i.p.) for 5 days consequently. Subsequent treatment with aqueous extract of CA (100 and 200 mg/kg) and standard drug methylcobalamin (MCA) (5 mg/kg) was done and results compared statistically. Neuropathic pain sensations were assessed using various behavioural and locomotory models and evaluated on 0th, 7th and 14th days. Kinovea software was used for video path-tracking of animals and total distance travelled calculated. The results indicated clear signs of improvement post 10 days of PT intoxication in CA-treated groups when compared PT challenged group. A significant reduction in pain behaviours in mechanical allodynia, cold chemical allodynia and thermal hyperalgesia models, improvement in sensory motor coordination, locomotor activity, and distance travelled in closed field model reveals that CA possesses potential ameliorating effect against PT-induced neuropathic pain symptoms. The extract notably improved the movement of the PT challenged animals which was shown by the video path-tracking software and total distance travelled by those animals.
Collapse
Affiliation(s)
- Varnita Karmakar
- Department of Pharmaceutical Sciences, Jharkhand Rai University, Ratu Road, Kamre, Ranchi, Jharkhand, 835222, India
| | - Firdous Sayeed Mohammad
- Department of Pharmacology, Calcutta Institute of Pharmaceutical Technology & AHS, Uluberia, Howrah, 711316, West Bengal, India.
| | - Saleh S Baeesa
- Division of Neurosurgery, College of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Athanasios Alexiou
- Novel Global Community Educational Foundation, Hebersham, Australia.,AFNP Med Austria, Wien, Austria
| | - S R Sivakumar
- Department of Botany, Bharathidasan University, Trichy, 620024, Tamilnadu, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia. .,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
482
|
Zhang XY, Vollert J, Sena ES, Rice AS, Soliman N. A protocol for the systematic review and meta-analysis of thigmotactic behaviour in the open field test in rodent models associated with persistent pain. BMJ OPEN SCIENCE 2022; 5:e100135. [PMID: 35047702 PMCID: PMC8647568 DOI: 10.1136/bmjos-2020-100135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/30/2020] [Accepted: 12/17/2020] [Indexed: 12/09/2022] Open
Abstract
Objective Thigmotaxis is an innate predator avoidance behaviour of rodents and is enhanced when animals are under stress. It is characterised by the preference of a rodent to seek shelter, rather than expose itself to the aversive open area. The behaviour has been proposed to be a measurable construct that can address the impact of pain on rodent behaviour. This systematic review will assess whether thigmotaxis can be influenced by experimental persistent pain and attenuated by pharmacological interventions in rodents. Search strategy We will conduct search on three electronic databases to identify studies in which thigmotaxis was used as an outcome measure contextualised to a rodent model associated with persistent pain. All studies published until the date of the search will be considered. Screening and annotation Two independent reviewers will screen studies based on the order of (1) titles and abstracts, and (2) full texts. Data management and reporting For meta-analysis, we will extract thigmotactic behavioural data and calculate effect sizes. Effect sizes will be combined using a random-effects model. We will assess heterogeneity and identify sources of heterogeneity. A risk-of-bias assessment will be conducted to evaluate study quality. Publication bias will be assessed using funnel plots, Egger’s regression and trim-and-fill analysis. We will also extract stimulus-evoked limb withdrawal data to assess its correlation with thigmotaxis in the same animals. The evidence obtained will provide a comprehensive understanding of the strengths and limitations of using thigmotactic outcome measure in animal pain research so that future experimental designs can be optimised. We will follow the Preferred Reporting Items for Systematic Reviews and Meta-Analyses reporting guidelines and disseminate the review findings through publication and conference presentation.
Collapse
Affiliation(s)
| | - Jan Vollert
- Musculoskeletal, Imperial College London, London, UK
| | - Emily S Sena
- Clinical Neurosciences, University of Edinburgh, Edinburgh, UK
| | | | - Nadia Soliman
- Musculoskeletal, Imperial College London, London, UK
| |
Collapse
|
483
|
de Abreu MS, Giacomini ACVV, Genario R, Demin KA, Amstislavskaya TG, Costa F, Rosemberg DB, Sneddon LU, Strekalova T, Soares MC, Kalueff AV. Understanding early-life pain and its effects on adult human and animal emotionality: Translational lessons from rodent and zebrafish models. Neurosci Lett 2022; 768:136382. [PMID: 34861343 DOI: 10.1016/j.neulet.2021.136382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 10/19/2022]
Abstract
Critical for organismal survival, pain evokes strong physiological and behavioral responses in various sentient species. Clinical and preclinical (animal) studies markedly increase our understanding of biological consequences of developmental (early-life) adversity, as well as acute and chronic pain. However, the long-term effects of early-life pain exposure on human and animal emotional responses remain poorly understood. Here, we discuss experimental models of nociception in rodents and zebrafish, and summarize mounting evidence of the role of early-life pain in shaping emotional traits later in life. We also call for further development of animal models to probe the impact of early-life pain exposure on behavioral traits, brain disorders and novel therapeutic treatments.
Collapse
Affiliation(s)
- Murilo S de Abreu
- Bioscreening Platform, School of Pharmacy, Southwest University, Chongqing, China; Bioscience Institute, University of Passo Fundo, Passo Fundo, RS, Brazil; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA.
| | - Ana C V V Giacomini
- Bioscience Institute, University of Passo Fundo, Passo Fundo, RS, Brazil; Postgraduate Program in Environmental Sciences, University of Passo Fundo, Passo Fundo, RS, Brazil
| | - Rafael Genario
- Bioscience Institute, University of Passo Fundo, Passo Fundo, RS, Brazil
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medcial Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Granov Russian Scientific Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Tamara G Amstislavskaya
- Scientific Research Institute of Neuroscience and Medicine, Novosibirsk, Russia; Novosibirsk State University, Novosibirsk, Russia; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Fabiano Costa
- Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Brazil; Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
| | - Denis B Rosemberg
- Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Lynne U Sneddon
- University of Gothenburg, Department of Biological and Environmental Sciences, Gothenburg, Sweden
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov 1st Moscow State Medical University, Moscow, Russia; Institute of General Pathology and Pathophysiology, Moscow, Russia; Department of Preventive Medicine, Maastricht Medical Center Annadal, Maastricht, Netherlands
| | - Marta C Soares
- CIBIO, Research Centre in Biodiversity and Genetic Resources, University of Porto, Campus Agrário de Vairão, Vairão, Portugal
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia.
| |
Collapse
|
484
|
Berke MS, Fensholdt LKD, Hestehave S, Kalliokoski O, Abelson KSP. Effects of buprenorphine on model development in an adjuvant-induced monoarthritis rat model. PLoS One 2022; 17:e0260356. [PMID: 35025864 PMCID: PMC8757907 DOI: 10.1371/journal.pone.0260356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 11/08/2021] [Indexed: 11/19/2022] Open
Abstract
Complete Freund’s adjuvant (CFA)-induced arthritis in rats is a common animal model for studying chronic inflammatory pain. However, modelling of the disease is associated with unnecessary pain and impaired animal wellbeing, particularly in the immediate post-induction phase. Few attempts have been made to counteract these adverse effects with analgesics. The present study investigated the effect of buprenorphine on animal welfare, pain-related behaviour and model-specific parameters during the disease progression in a rat model of CFA-induced monoarthritis. The aim was to reduce or eliminate unnecessary pain in this model, in order to improve animal welfare and to avoid suffering, without compromising the quality of the model. Twenty-four male Sprague Dawley rats were injected with 20 μl of CFA into the left tibio-tarsal joint to induce monoarthritis. Rats were treated with either buprenorphine or carprofen for 15 days during the disease development, and were compared to a saline-treated CFA-injected group or a negative control group. Measurements of welfare, pain-related behaviour and clinical model-specific parameters were collected. The study was terminated after 3 weeks, ending with a histopathologic analysis. Regardless of treatment, CFA-injected rats displayed mechanical hyperalgesia and developed severe histopathological changes associated with arthritis. However, no severe effects on general welfare were found at any time. Buprenorphine treatment reduced facial pain expression scores, improved mobility, stance and lameness scores and it did not supress the CFA-induced ankle swelling, contrary to carprofen. Although buprenorphine failed to demonstrate a robust analgesic effect on the mechanical hyperalgesia in this study, it did not interfere with the development of the intended pathology.
Collapse
Affiliation(s)
- Mie S Berke
- Dept. of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Louise K D Fensholdt
- Dept. of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sara Hestehave
- Dept. of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Otto Kalliokoski
- Dept. of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Klas S P Abelson
- Dept. of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
485
|
Drevet S, Favier B, Brun E, Gavazzi G, Lardy B. Mouse Models of Osteoarthritis: A Summary of Models and Outcomes Assessment. Comp Med 2022; 72:3-13. [PMID: 34986927 DOI: 10.30802/aalas-cm-21-000043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Osteoarthritis (OA) is a multidimensional health problem and a common chronic disease. It has a substantial impact onpatient quality of life and is a common cause of pain and mobility issues in older adults. The functional limitations, lack of curative treatments, and cost to society all demonstrate the need for translational and clinical research. The use of OA models in mice is important for achieving a better understanding of the disease. Models with clinical relevance are needed to achieve 2 main goals: to assess the impact of the OA disease (pain and function) and to study the efficacy of potential treatments. However, few OA models include practical strategies for functional assessment of the mice. OA signs in mice incorporate complex interrelations between pain and dysfunction. The current review provides a comprehensive compilation of mousemodels of OA and animal evaluations that include static and dynamic clinical assessment of the mice, merging evaluationof pain and function by using automatic and noninvasive techniques. These new techniques allow simultaneous recordingof spontaneous activity from thousands of home cages and also monitor environment conditions. Technologies such as videographyand computational approaches can also be used to improve pain assessment in rodents but these new tools must first be validated experimentally. An example of a new tool is the digital ventilated cage, which is an automated home-cage monitor that records spontaneous activity in the cages.
Collapse
|
486
|
Wang Y, Liu Y, Liu J, Wang M, Wang Y. Coadministration of Curcumin and Hydromorphone Hydrochloride Alleviates Postoperative Pain in Rats. Biol Pharm Bull 2022; 45:27-33. [PMID: 34980778 DOI: 10.1248/bpb.b21-00434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study aimed to explore the effect of curcumin and hydromorphone hydrochloride (HH) cotreatment on postoperative pain in rats. An incision + formaldehyde-induced pain rat model was established. Rats were treated with vehicle, curcumin, HH, or curcumin + HH. Paw mechanical withdrawal threshold and thermal withdrawal latency were measured at 1 d before surgery as well as 1 , 2 h, 1 , 3 , and 7 d after surgery to assess pain sensitivity. The L4-6 region of the spinal cord was collected from each rat at 2 h, 1 , 3 , and 7 d after surgery. Western blot analysis and immunohistochemical staining were carried out to detect the protein expression of pain-related genes. Quantitative real-time PCR and enzyme-linked immunosorbent assay were conducted to measure the expression and production of proinflammatory mediators. Compared with other groups, Curcumin + HH significantly reduced pain sensitivity in the model rats. Mechanistically, curcumin + HH suppressed protein expression of stromal cell-derived factor-1 (SDF-1), CXC chemokine receptor 4 (CXCR4), p-Akt, and c-fos while enhancing protein expression of nerve growth factor (NGF) in the dorsal root ganglia (DRG) of model rats. Curcumin + HH inhibited the expression and production of interleukin 1β (IL-1β), cyclooxygenase-2 (COX-2), tumor necrosis factor α (TNF-α), and p65 nuclear factor kappa B (NF-κB) in the DRG. Coadministration of curcumin and HH alleviates incision + formaldehyde-induced pain in rats, possibly by suppressing the SDF-1/CXCR4 pathway and the production of proinflammatory mediators. Our results provide curcumin and HH cotreatment as a promising therapeutic strategy in the management of postoperative pain.
Collapse
Affiliation(s)
- Yihan Wang
- Department of anesthesiology, Lanzhou University Second Hospital
| | - Yang Liu
- Department of anesthesiology, Lanzhou University Second Hospital
| | - Jieting Liu
- Department of anesthesiology, Lanzhou University Second Hospital
| | - Min Wang
- Department of anesthesiology, Lanzhou University Second Hospital
| | - Yingbin Wang
- Department of anesthesiology, Lanzhou University Second Hospital
| |
Collapse
|
487
|
Morgese MG, Bove M, Di Cesare Mannelli L, Schiavone S, Colia AL, Dimonte S, Mhillaj E, Sikora V, Tucci P, Ghelardini C, Trabace L. Precision Medicine in Alzheimer's Disease: Investigating Comorbid Common Biological Substrates in the Rat Model of Amyloid Beta-Induced Toxicity. Front Pharmacol 2022; 12:799561. [PMID: 35046821 PMCID: PMC8763383 DOI: 10.3389/fphar.2021.799561] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/01/2021] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD), one of the most widespread neurodegenerative disorder, is a fatal global burden for the elder population. Although many efforts have been made, the search of a curative therapy is still ongoing. Individuating phenotypic traits that might help in investigating treatment response is of growing interest in AD research. AD is a complex pathology characterized by many comorbidities, such as depression and increased susceptibility to pain perception, leading to postulate that these conditions may rely on common biological substrates yet to be determined. In order to investigate those biological determinants to be associable with phenotypic traits, we used the rat model of amyloid beta-induced toxicity. This established model of early phase of AD is obtained by the intracerebroventricular injection of soluble amyloid beta1-42 (Aβ) peptide 7 days before performing experiments. In this model, we have previously reported increased immobility in the forced swimming test, reduced cortical serotonin levels and subtle alterations in the cognitive domain a depressive-like phenotype associated with subtle alteration in memory processes. In light of evaluating pain perception in this animal model, we performed two different behavioral tests commonly used, such as the paw pressure test and the cold plate test, to analyze mechanical hyperalgesia and thermal allodynia, respectively. Behavioural outcomes confirmed the memory impairment in the social recognition test and, compared to sham, Aβ-injected rats showed an increased selective susceptibility to mechanical but not to thermal stimulus. Behavioural data were then corroborated by neurochemical and biochemical biomarker analyses either at central or peripheral level. Data showed that the peptide injection evoked a significant increase in hypothalamic glutamate, kynurenine and dopamine content, while serotonin levels were reduced. Plasma Cystatin-C, a cysteine protease, was increased while serotonin and melatonin levels were decreased in Aβ-injected rats. Urinary levels paralleled plasma quantifications, indicating that Aβ-induced deficits in pain perception, mood and cognitive domain may also depend on these biomarkers. In conclusion, in the present study, we demonstrated that this animal model can mimic several comorbid conditions typical of the early phase of AD. Therefore, in the perspective of generating novel therapeutic strategies relevant to precision medicine in AD, this animal model and the biomarkers evaluated herein may represent an advantageous approach.
Collapse
Affiliation(s)
- Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria Bove
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Lorenzo Di Cesare Mannelli
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Firenze, Firenze, Italy
| | - Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Anna Laura Colia
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Stefania Dimonte
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Emanuela Mhillaj
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Vladyslav Sikora
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
- Department of Pathology, Sumy State University, Sumy, Ukraine
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Carla Ghelardini
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Firenze, Firenze, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
488
|
The effect of tacrolimus-containing polyethylene glycol-modified maghemite nanospheres on reducing oxidative stress and accelerating the healing spinal cord injury of rats based on increasing M2 macrophages. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2021.103534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
489
|
Estrázulas M, Freitas RDS, Käfer ET, Dagnino APA, Campos MM. Central and peripheral effects of environmental enrichment in a mouse model of arthritis. Int Immunopharmacol 2022; 102:108386. [PMID: 34824037 DOI: 10.1016/j.intimp.2021.108386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/14/2021] [Accepted: 11/17/2021] [Indexed: 01/07/2023]
Abstract
This study analyzed whether environmental enrichment (EE) modulates the nociceptive and inflammatory responses in the mouse model of arthritis induced by Complete Freund's Adjuvant (CFA). Ninety male mice (C57BL/6-JUnib, 4-weeks-old; 20-25 g) were distributed into EE and standard (SE) groups. For EE, mice were kept in bigger cages using an alternation of materials to chew (wood and paper), for nesting (cotton), to use as hiding places (plastic tunnels), and for voluntary exercise (wheel running). Arthritis was induced by an injection of CFA (50 μL) into the right hind paw or saline solution in the control group. Separate groups received the anti-inflammatory drug dexamethasone (0.5 mg/kg; every 48 h). Inflammatory and pain measurements were performed from 1 to 35 days after CFA administration. EE per se reduced the acute paw edema formation and arthritis scores. The serum levels of tumor necrosis factor (TNF) were undetectable in any experimental groups. EE diminished the immunopositivity for the microglia marker IBA1 in the pre-frontal cortex, with slight changes for hippocampal GFAP-positive activated astrocytes. Finally, EE induced a marked increment of brain-derived nerve factor (BDNF) expression in the hippocampus, an effect that was fully prevented by dexamethasone. These data bring novel evidence on the peripheral and central effects of EE in a mouse arthritis model.
Collapse
Affiliation(s)
- Marina Estrázulas
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Centro de Pesquisas em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Raquel D S Freitas
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Centro de Pesquisas em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Eduarda T Käfer
- Curso de Graduação em Medicina, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ana P A Dagnino
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Centro de Pesquisas em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria M Campos
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Centro de Pesquisas em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
490
|
Green-Fulgham SM, Ball JB, Maier SF, Rice KC, Watkins LR, Grace PM. Suppression of active phase voluntary wheel running in male rats by unilateral chronic constriction injury: Enduring therapeutic effects of a brief treatment of morphine combined with TLR4 or P2X7 antagonists. J Neurosci Res 2022; 100:265-277. [PMID: 32533604 PMCID: PMC8377614 DOI: 10.1002/jnr.24645] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/21/2020] [Accepted: 05/03/2020] [Indexed: 12/20/2022]
Abstract
The present series of studies examine the impact of systemically administered therapeutics on peripheral nerve injury (males; unilateral sciatic chronic constriction injury [CCI])-induced suppression of voluntary wheel running, across weeks after dosing cessation. Following CCI, active phase running distance and speed are suppressed throughout the 7-week observation period. A brief course of morphine, however, increased active phase running distance and speed throughout this same period, an effect apparent only in sham rats. For CCI rats, systemic co-administration of morphine with antagonists of either P2X7 (A438079) or TLR4 ((+)-naloxone) (receptors critical to the activation of NLRP3 inflammasomes and consequent inflammatory cascades) returned running behavior of CCI rats to that of shams through 5+ weeks after dosing ceased. This is a striking difference in effect compared to our prior CCI allodynia results using systemic morphine plus intrathecal delivery of these same antagonists, wherein a sustained albeit partial suppression of neuropathic pain was observed. This may point to actions of the systemic drugs at multiple sites along the neuraxis, modulating injury-induced, inflammasome-mediated effects at the injured sciatic nerve and/or dorsal root ganglia, spinal cord, and potentially higher levels. Given that our data to date point to morphine amplifying neuroinflammatory processes put into motion by nerve injury, it is intriguing to speculate that co-administration of TLR4 and/or P2X7 antagonists can intervene in these inflammatory processes in a beneficial way. That is, that systemic administration of such compounds may suppress inflammatory damage at multiple sites, rapidly and persistently returning neuropathic animals to sham levels of response.
Collapse
Affiliation(s)
- Suzanne M. Green-Fulgham
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Jayson B. Ball
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Steven F. Maier
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Kenner C. Rice
- Drug Design and Synthesis Section, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, Bethesda, MD, USA
| | - Linda R. Watkins
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Peter M. Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
491
|
Li YX, Li JH, Guo Y, Tao ZY, Qin SH, Traub RJ, An H, Cao DY. Oxytocin inhibits hindpaw hyperalgesia induced by orofacial inflammation combined with stress. Mol Pain 2022; 18:17448069221089591. [PMID: 35266833 PMCID: PMC9047792 DOI: 10.1177/17448069221089591] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Oxytocin (OT) is recognized as a critical neuropeptide in pain-related disorders. Chronic pain caused by the comorbidity of temporomandibular disorder (TMD) and fibromyalgia syndrome (FMS) is common, but whether OT plays an analgesic role in the comorbidity of TMD and FMS is unknown. Female rats with masseter muscle inflammation combined with 3-day forced swim (FS) stress developed somatic hypersensitivity, which modeled the comorbidity of TMD and FMS. Using this model, the effects of spinal OT administration on mechanical allodynia and thermal hyperalgesia in hindpaws were examined. Furthermore, the protein levels of OT receptors and 5-HT2A receptors in the L4-L5 spinal dorsal horn were analyzed by Western blot. The OT receptor antagonist atosiban and 5-HT2A receptor antagonist ritanserin were intrathecally injected prior to OT injection in the separate groups. Intrathecal injection of 0.125 μg and 0.5 μg OT attenuated the hindpaw hyperalgesia. The expression of OT receptors and 5-HT2A receptors in the L4-L5 spinal dorsal horn significantly increased following intrathecal injection of 0.5 μg OT. Intrathecal administration of either the OT receptor antagonist atosiban or 5-HT2A receptor antagonist ritanserin blocked the analgesic effect of OT. These results suggest that OT may inhibit hindpaw hyperalgesia evoked by orofacial inflammation combined with stress through OT receptors and/or 5-HT2A receptors, thus providing a therapeutic prospect for drugs targeting the OT system and for patients with comorbidity of TMD and FMS.
Collapse
Affiliation(s)
- Yue-Xin Li
- Key Laboratory of Shaanxi Province
for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi’an Jiaotong University College of
Stomatology, China
- Department of Special Dental Care, Xi’an Jiaotong University College of
Stomatology, China
| | - Jia-Heng Li
- Key Laboratory of Shaanxi Province
for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi’an Jiaotong University College of
Stomatology, China
| | - Yi Guo
- Key Laboratory of Shaanxi Province
for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi’an Jiaotong University College of
Stomatology, China
| | - Zhuo-Ying Tao
- Key Laboratory of Shaanxi Province
for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi’an Jiaotong University College of
Stomatology, China
| | - Shi-Hao Qin
- Key Laboratory of Shaanxi Province
for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi’an Jiaotong University College of
Stomatology, China
- Department of Special Dental Care, Xi’an Jiaotong University College of
Stomatology, China
| | - Richard J Traub
- Department of Neural and Pain
Sciences, School of Dentistry, Center to Advance Chronic Pain Research, University of Maryland
Baltimore, Baltimore, MD, USA
| | - Hong An
- Department of Special Dental Care, Xi’an Jiaotong University College of
Stomatology, China
| | - Dong-Yuan Cao
- Key Laboratory of Shaanxi Province
for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi’an Jiaotong University College of
Stomatology, China
| |
Collapse
|
492
|
Methods and protocols for chemotherapy-induced peripheral neuropathy (CIPN) mouse models using paclitaxel. Methods Cell Biol 2022; 168:277-298. [DOI: 10.1016/bs.mcb.2021.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
493
|
Leahy T, Nuss C, Evans MK, Fung A, Shetye S, Soslowsky LJ. Achilles Tendon Ruptures in Middle-Aged Rats Heal Poorly Compared With Those in Young and Old Rats. Am J Sports Med 2022; 50:170-181. [PMID: 34851182 PMCID: PMC8819270 DOI: 10.1177/03635465211055476] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Achilles tendon ruptures are painful and debilitating injuries and are most common in middle-aged patients. There is a lack of understanding of the underlying causes for increased rupture rates in middle-aged patients and how healing outcomes after a rupture might be affected by patient age. Therefore, the objective of this study was to define age-specific Achilles tendon healing by assessing ankle functional outcomes and Achilles tendon mechanical and histological properties after a rupture using a rat model. HYPOTHESIS Rats representing the middle-aged patient population would demonstrate reduced healing capability after an Achilles tendon rupture, as demonstrated by a slower return to baseline ankle functional properties and inferior biomechanical and histological tendon properties. STUDY DESIGN Controlled laboratory study. METHODS Fischer 344 rats were categorized by age to represent young, middle-aged, and old patients, and Achilles tendon ruptures were induced in the right hindlimb. Animals were allowed to heal and were euthanized at 3 or 6 weeks after the injury. In vivo functional assays and ultrasound imaging were performed throughout the healing period, and ex vivo tendon mechanical and histological properties were assessed after euthanasia. RESULTS Rats representing middle-aged patients displayed reduced healing potential compared with the other age groups, as they demonstrated decreased recovery of in vivo functional and ultrasound assessment parameters and inferior mechanical and histological properties after an Achilles tendon rupture. CONCLUSION These findings may help explain the increased rupture rate observed clinically in middle-aged patients by suggesting that there may be altered tendon responses to daily trauma. CLINICAL RELEVANCE The results provide novel data on age-specific healing outcomes after an Achilles tendon rupture, which underscores the importance of considering a patient's age during treatment and expectations for outcomes.
Collapse
Affiliation(s)
- Thomas Leahy
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Courtney Nuss
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary Kate Evans
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley Fung
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Snehal Shetye
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Louis J. Soslowsky
- McKay Orthopaedic Laboratory, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
494
|
Do W, Baik J, Jeon S, You CM, Kang D, Jung YH, Lee J, Kim HK. Increased Brain-Derived Neurotrophic Factor Levels in Cerebrospinal Fluid During the Acute Phase in TBI-Induced Mechanical Allodynia in the Rat Model. J Pain Res 2022; 15:229-239. [PMID: 35125890 PMCID: PMC8809523 DOI: 10.2147/jpr.s344110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/21/2022] [Indexed: 11/23/2022] Open
Abstract
Background The present study aimed to develop a rat model for mechanical allodynia after traumatic brain injury (TBI) and to investigate the expression of brain-derived neurotrophic factor (BDNF) in the cerebrospinal fluid (CSF) using this model. Methods A total of 180 rats were randomly allocated into three groups: a control group (group C), a sham-operated group (group S), and a controlled cortical impact induced TBI group (group T), 60 in each group. Von Frey test was performed to evaluate mechanical withdrawal thresholds. An enzyme-linked immunosorbent assay was performed to quantify BDNF level in CSF. Results The 50% withdrawal thresholds of group T were lower than those of group C and group S at all measuring points except for the preoperative period (P = 0.026, <0.001, and <0.001 for POD1, POD7, and POD14, respectively). The BDNF level of group T was higher than those of group C and group S at POD1 (P = 0.005). Conclusion Upregulation of the BDNF expression in CSF was observed in rats who developed mechanical allodynia on the day after TBI. Based on our findings, to elucidate the relationship between TBI-induced neuropathic pain and BDNF expression in CSF, further research should be carried out through a multifaceted approach to a broad spectrum of pain behavior models.
Collapse
Affiliation(s)
- Wangseok Do
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
| | - Jiseok Baik
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, Republic of Korea
- Correspondence: Jiseok Baik, Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, 179 Gudeok-Ro, Seo-gu, Busan, 49241, Republic of Korea, Tel +82-51-240-7499, Fax +82-51-242-7466, Email
| | - Soeun Jeon
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
| | - Chang-Min You
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
| | - Dahyun Kang
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
| | - Young-Hoon Jung
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
| | - Jiyoon Lee
- Department of Anesthesia and Pain Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hae-Kyu Kim
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
495
|
Dorval L, Knapp BI, Majekodunmi OA, Eliseeva S, Bidlack JM. Mice with high FGF21 serum levels had a reduced preference for morphine and an attenuated development of acute antinociceptive tolerance and physical dependence. Neuropharmacology 2022; 202:108858. [PMID: 34715121 PMCID: PMC8627472 DOI: 10.1016/j.neuropharm.2021.108858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 10/18/2021] [Accepted: 10/23/2021] [Indexed: 01/03/2023]
Abstract
Because of increased opioid misuse, there is a need to identify new targets for minimizing opioid tolerance, and physical and psychological dependence. Previous studies showed that fibroblast growth factor 21 (FGF21) decreased alcohol and sweet preference in mice. In this study, FGF21-transgenic (FGF21-Tg) mice, expressing high FGF21 serum levels, and wildtype (WT) C57BL/6J littermates were treated with morphine and saline to determine if differences exist in their physiological and behavioral responses to opioids. FGF21-Tg mice displayed reduced preference for morphine in the conditioned place preference assay compared to WT littermates. Similarly, FGF21-Tg mice had an attenuation of the magnitude and rate of acute morphine antinociceptive tolerance development, and acute and chronic morphine physical dependence, but exhibited no change in chronic morphine antinociceptive tolerance. The ED50 values for morphine-induced antinociception in the 55 °C hot plate and the 55 °C warm-water tail withdrawal assays were similar in both strains of mice. Likewise, FGF21-Tg and WT littermates had comparable responses to morphine-induced respiratory depression. Overall, FGF21-Tg mice had a decrease in the development of acute analgesic tolerance, and the development of physical dependence, and morphine preference. FGF21 and its receptor have therapeutic potential for reducing opioid withdrawal symptoms and craving, and augmenting opioid therapeutics for acute pain patients to minimize tolerance development.
Collapse
Affiliation(s)
- Louben Dorval
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, 14642, Rochester, NY, USA
| | - Brian I Knapp
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, 14642, Rochester, NY, USA
| | - Olufolake A Majekodunmi
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, 14642, Rochester, NY, USA
| | - Sophia Eliseeva
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, School of Medicine and Dentistry, 14642, Rochester, NY, USA
| | - Jean M Bidlack
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, 14642, Rochester, NY, USA.
| |
Collapse
|
496
|
Vasudevan R, Kandasamy G, Almaghaslah D, Almanasef M, Alqahatani A, Aldahish A, Venkatesan K, Paulsamy P, Maheswari C, Wahab S. Alleviation of neuropathic pain by trazodone in rats. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
497
|
Methods and protocols for translatable rodent models of postsurgical pain. Methods Cell Biol 2022; 168:249-276. [DOI: 10.1016/bs.mcb.2021.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
498
|
Radulescu A, White FA, Chenu C. What Did We Learn About Fracture Pain from Animal Models? J Pain Res 2022; 15:2845-2856. [PMID: 36124034 PMCID: PMC9482434 DOI: 10.2147/jpr.s361826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 07/01/2022] [Indexed: 11/23/2022] Open
Abstract
Progress in bone fracture repair research has been made possible due to the development of reproducible models of fracture in rodents with more clinically relevant fracture fixation, where there is considerably better assessment of the factors that affect fracture healing and/or novel therapeutics. However, chronic or persistent pain is one of the worst, longest-lasting and most difficult symptoms to manage after fracture repair, and an ongoing challenge remains for animal welfare as limited information exists regarding pain scoring and management in these rodent fracture models. This failure of adequate pre-clinical pain assessment following osteotomy in the rodent population may not only subject the animal to severe pain states but may also affect the outcome of the bone healing study. Animal models to study pain were also mainly developed in rodents, and there is increasing validation of fracture and pain models to quantitatively evaluate fracture pain and to study the factors that generate and maintain fracture pain and develop new therapies for treating fracture pain. This review aims to discuss the different animal models for fracture pain research and characterize what can be learned from using animal models of fracture regarding behavioral pain states and new molecular targets for future management of these behaviors.
Collapse
Affiliation(s)
- Andreea Radulescu
- Royal Veterinary College, Department of Comparative Biomedical Sciences, London, NW1 OTU, UK
| | - Fletcher A White
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush Veterans Medical Center, Indianapolis, IN, USA
| | - Chantal Chenu
- Royal Veterinary College, Department of Comparative Biomedical Sciences, London, NW1 OTU, UK
- Correspondence: Chantal Chenu, Royal Veterinary College, Department of Comparative Biological Sciences, Royal College Street, London, NW1 0TU, UK, Tel +44 207 468 5045, Email
| |
Collapse
|
499
|
Kao AR, Xu C, Gerling GJ. Using Digital Image Correlation to Quantify Skin Deformation With Von Frey Monofilaments. IEEE TRANSACTIONS ON HAPTICS 2022; 15:26-31. [PMID: 34951855 PMCID: PMC9006180 DOI: 10.1109/toh.2021.3138350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Thin von Frey monofilaments are a clinical tool used worldwide to assess touch deficits. One's ability to perceive touch with low-force monofilaments (0.008 - 0.07 g) establishes an absolute threshold and thereby the extent of impairment. While individual monofilaments bend at defined forces, there are no empirical measurements of the skin surface's response. In this work, we measure skin surface deformation at light-touch perceptual limits, by adopting an imaging approach using 3D digital image correlation (DIC). Generating point cloud data from three cameras surveilling the index finger pad, we reassemble and stitch together multiple 3D surfaces. Then, in response to each monofilament's indentation over time, we quantify strain across the skin surface, radial deformation emanating from the contact point, penetration depth into the surface, and area between 2D cross-sections. The results show that the monofilaments create distinct states of skin deformation, which align closely with just noticeable percepts at absolute detection and discrimination thresholds, even amidst variance between individuals and trials. In particular, the resolution of the DIC imaging approach captures sufficient differences in skin deformation at threshold, offering promise in understanding the skin's role in perception.
Collapse
|
500
|
Dyachenko IA, Palikova YA, Palikov VA, Korolkova YV, Kazakov VA, Egorova NS, Garifulina AI, Utkin YN, Tsetlin VI, Kryukova EV. α-Conotoxin RgIA and oligoarginine R8 in the mice model alleviate long-term oxaliplatin induced neuropathy. Biochimie 2021; 194:127-136. [PMID: 34979156 DOI: 10.1016/j.biochi.2021.12.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/28/2022]
Abstract
Оligoarginines were recently discovered (Lebedev et al., 2019 Nov) [1] as a novel class of nicotinic acetylcholine receptors (nAChRs) inhibitors, octaoligoarginine R8 showing a relatively high affinity (40 nM) for the α9/α10 nAChR. Since the inhibition of α9/α10 nAChR by α-conotoxin RgIA and its analogs is a possible way to drugs against neuropathic pain, here in a mice model we compared R8 with α-conotoxin RgIA in the effects on the chemotherapy-induced peripheral neuropathy (CIPN), namely on the long-term oxaliplatin induced neuropathy. Tests of cold allodynia, hot plate, Von Frey and grip strength analysis revealed for R8 and α-conotoxin RgIA similar positive effects, expressed most prominently after two weeks of administration. Histological analysis of the dorsal root ganglia sections showed for R8 and RgIA a similar partial correction of changes in the nuclear morphology of neurons. Since α9/α10 nAChR might be not the only drug target for R8, we analyzed the R8 action on rat TRPV1 and TRPA1, well-known nociceptive receptors. Against rTRPV1 at 25 μM there was no inhibition, while for rTRPA1 IC50 was about 20 μM. Thus, involvement of rTRPA1 cannot be excluded, but in view of the R8 much higher affinity for α9/α10 nAChR the latter seems to be the main target and the easily synthesized R8 can be considered as a potential candidate for a drug design.
Collapse
Affiliation(s)
- I A Dyachenko
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290, Pushchino, Moscow, Russia.
| | - Yu A Palikova
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290, Pushchino, Moscow, Russia.
| | - V A Palikov
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290, Pushchino, Moscow, Russia.
| | - Y V Korolkova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| | - V A Kazakov
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290, Pushchino, Moscow, Russia.
| | - N S Egorova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| | - A I Garifulina
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, A-1090, Vienna, Austria.
| | - Y N Utkin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| | - V I Tsetlin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| | - E V Kryukova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| |
Collapse
|