Next Article in Journal
Job Analysis and Curriculum Design of South Korean Animal-Assisted Therapy Specialists Using DACUM
Previous Article in Journal
Motivation and Habits of a Wild Boar-Hunting Community
Previous Article in Special Issue
Faecal Short-Chain, Long-Chain, and Branched-Chain Fatty Acids as Markers of Different Chronic Inflammatory Enteropathies in Dogs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Evaluation of the Therapeutic Potential of Amantadine in a Vincristine-Induced Peripheral Neuropathy Model in Rats

by
Isabela Santana Albertazzi Drummond
1,
Jéssica Natália Silva de Oliveira
1,
Raquel Vieira Niella
1,
Álvaro José Chávez Silva
1,
Iago Santos de Oliveira
1,
Sophia Saraiva de Souza
1,
Claire Souza da Costa Marques
1,
Janaina Maria Xavier Corrêa
1,
Juneo Freitas Silva
2 and
Mário Sérgio Lima de Lavor
1,*
1
Department of Agricultural and Environmental Sciences, State University of Santa Cruz (UESC), Ilhéus 45662-900, BA, Brazil
2
Department of Biological Sciences, State University of Santa Cruz (UESC), Ilhéus 45662-900, BA, Brazil
*
Author to whom correspondence should be addressed.
Animals 2024, 14(13), 1941; https://doi.org/10.3390/ani14131941
Submission received: 20 May 2024 / Revised: 24 June 2024 / Accepted: 25 June 2024 / Published: 30 June 2024
(This article belongs to the Special Issue Advanced Biomarker Research in Animal Pathological States)

Abstract

:

Simple Summary

Vincristine-induced peripheral neuropathy is a debilitating side effect and a limiting factor in the continuance of treatment of cancer patients. Thus, therapeutic strategies are necessary to enable the maintenance of oncological treatment and minimize undesired conditions caused by the treatment. Current protocols for the management of neuropathic pain remain unsatisfactory due to the complexity and severity of clinical signs, as well as the lack of significant improvement with pharmacological treatment. Amantadine shows therapeutic potential by acting on NMDA receptors. Immunohistochemistry, quantitative PCR and analysis of enzymatic activity were performed to demonstrate the therapeutic activity of amantadine in the spinal cord of rats with induced neuropathic pain. The authors herein demonstrated positive effects on the regulation of neuroinflammation, oxidative stress, reticulum endoplasmic stress and apoptosis with the highest doses examined (25 mg/kg and 50 mg/kg). We hope that our research will contribute to studies of amantadine as an option in a pharmacotherapeutic protocol for peripheral neuropathy.

Abstract

This study aimed to evaluate the therapeutic potential of amantadine in a vincristine-induced peripheral neuropathy model in rats. Forty-eight male Wistar rats were used. The treated groups received oral amantadine at doses of 2, 5, 12, 25 and 50 mg/kg, with daily applications for 14 days. The mechanical paw withdrawal threshold was measured using a digital analgesimeter. Immunohistochemical analysis of IL-6, TNFα, MIP1α, IL-10, CX3CR1, CXCR4, SOD, CAT and GPx, and enzymatic activity analysis of CAT, SOD and GPx were performed, in addition to quantitative PCR of Grp78, Chop, Ho1, Perk, Bax, Bcl-xL, Casp 3, Casp 9, IL-6, IL-10, IL-18 and IL-1β. The results showed an increase in nociceptive thresholds in animals that received 25 mg/kg and 50 mg/kg amantadine. Immunohistochemistry showed a decrease in the immunostaining of IL-6, TNFα, MIP1α and CX3CR1, and an increase in IL-10. CAT and SOD showed an increase in both immunochemistry and enzymatic analysis. qPCR revealed a reduced expression of genes related to endoplasmic reticulum stress and regulation in the expression of immunological and apoptotic markers. Amantadine demonstrated antinociceptive, anti-inflammatory and antioxidant effects in the vincristine-induced peripheral neuropathy model in rats, suggesting that amantadine may be considered an alternative approach for the treatment of vincristine-induced peripheral neuropathic pain.

1. Introduction

Cancer is one of the main causes of decreased quality of life worldwide, with a high death rate [1]. In the last global estimate carried out by Globocan in 2020, it was estimated that around 19.3 million new cases of cancer were identified in 2020 alone and approximately 10 million cancer deaths in that same year [2,3]. The World Health Organization, in 2019, estimated that, in 112 countries, cancer was the first or second leading cause of death for people under 70 years of age, and in a further 23 countries it ranked third or fourth [4]. In the treatment of cancer patients, chemotherapy is one of the methods used, in addition to radiotherapy, surgery and supportive treatments. Chemotherapy drugs routinely used in the clinic are not pharmacologically selective, which means that they interact non-specifically on tumor and healthy cells, causing adverse effects on the latter [5].
Among chemotherapy drugs of great importance in medicine, vincristine is used in several protocols for various cancers, such as acute leukemia, Hodgkin’s disease, neuroblastoma, uterine cervix carcinoma, lymphomas, breast cancer and melanoma, and is also used in therapeutic protocols for other neoplasms in association with additional chemotherapy agents [6]. The most commonly observed adverse effects induced by this drug include peripheral neuropathy, paresthesia, gastrointestinal tract disorders, namely anorexia, diarrhea and vomiting, alopecia and mild immunosuppression [7]. Most patients treated with vincristine with a cumulative dose greater than 4 mg/m2 develop some degree of sensory peripheral neuropathy [8].
Peripheral neuropathy is the most common neurological syndrome secondary to antineoplastic therapy, and occurs in a dose-dependent manner, which can disrupt the chemotherapy protocol by leading to drug changes, dose reductions and even discontinuation of treatment [9]. Of cancer patients undergoing treatment, more than 60% have peripheral neuropathy linked to the use of chemotherapy drugs, with chemotherapy-induced peripheral neuropathy (CIPN) being one of the most common adverse effects of chemotherapy [10]. Its pathophysiology is multifactorial and involves neuroinflammation, immunological processes, oxidative stress, apoptotic mechanisms, changes in calcium homeostasis, axon degeneration and membrane remodeling [11].
The treatment of neuropathic pain is still uncertain and is still far from satisfactory, as most of those patients do not achieve significant improvement with pharmacological treatment, and the clinical signs of neuropathic pain can be severe and respond poorly to analgesic therapeutic protocols [12]. Opioids were used as first-line agents, but they still did not reach an attractive therapeutic significance [13]. Soon after, anticonvulsants were added to the class of first-line medications, but their effectiveness was still limited. Currently, non-steroidal anti-inflammatory drugs (NSAIDs), alpha-2-adrenergic agonists, antidepressants and NMDA antagonists are most commonly used. Studies suggest interest in the therapeutic efficacy in neuropathic pain of N-methyl-D-aspartate (NMDA) receptor antagonists [14,15,16].
Amantadine, a drug initially used as an antiviral and for the treatment of the influenza virus and in the treatment of Parkinson’s, is also being considered for pain management, since its mechanism of action promotes the non-competitive antagonism of glutamatergic receptor damage of the NMDA type, which participates in the neurophysiological mechanism of nociception [17].
Although the literature on the use of amantadine in relieving neuropathic pain is scarce, studies suggest its effectiveness in modulating central sensitization [18,19]. Amantadine promoted a reduction in oxidative stress and prevented hypersensitivity in spinal cord injury in rats [20]. In cats, the combination of meloxicam and amantadine ensured clinical improvement in sciatic nerve injury [21].
In accordance with the need for a major understanding of the efficacy of amantadine in oncological neuropathy, this study aimed to clarify the antinociceptive potential of amantadine through the assessment of the mechanical nociceptive threshold, in addition to elucidating its inhibitory effect on the production of reactive oxygen species, lipid peroxidation and enzymatic activity and evaluating its anti-inflammatory effect on a vincristine-induced neuropathy model.

2. Materials and Methods

2.1. Animals and Housing Conditions

Forty-eight male Wistar rats (Rattus norvegicus) were used, weight ranging from 250–300 g, from the Laboratory Animal Rearing and Maintenance Station of the State University of Santa Cruz–UESC, Ilhéus, BA, Brazil. The animals were kept in the Experimental Laboratory of the UESC Veterinary Hospital in 1500 cm2 polypropylene boxes lined with wood shavings (containing five animals each) in a temperature-controlled environment (22 °C) and a 12 h photoperiod (light/dark), receiving commercial food and water ad libitum, with an acclimation period to the experimental conditions of one week, to equalize all environmental and food conditions, and reduce possible interference with the research results.

2.2. Chemotherapy-Induced Neuropathic Pain Model and Experimental Groups

Vincristine was administered at doses of 0.05 mg/kg/day intraperitoneally (IP), once a day for 14 days, to develop the experimental neuropathic pain model [22]. The negative control group (GCN) received saline solution (0.9% NaCl) intraperitoneally in a volume of 0.1 mL/animal, once a day for 14 days. Animals treated with amantadine received oral amantadine in different doses (2 mg, 5 mg, 12 mg, 25 mg or 50 mg), once a day for 14 days, one hour before the administration of vincristine sulfate.
The animals were randomly divided into eight groups (n = 6):
  • Naïve group (GN): animals did not receive any drug or vehicle;
  • Negative control group (GCN): saline solution only (NaCl 0.9%);
  • Positive control group (GCP): vincristine sulfate only 0.05 mg/kg/day;
  • Group GA2: amantadine 2 mg/kg/day + vincristine sulfate 0.05 mg/kg/day;
  • Group GA5: amantadine 5 mg/kg/day + vincristine sulfate 0.05 mg/kg/day;
  • Group GA12: amantadine 12 mg/kg/day + vincristine sulfate 0.05 mg/kg/day;
  • Group GA25: amantadine 25 mg/kg/day + vincristine sulfate 0.05 mg/kg/day;
  • Group GA50: amantadine 50 mg/kg/day + vincristine sulfate 0.05 mg/kg/day.

2.3. Assessment of Mechanical Nociceptive Threshold

Assessment of nociceptive mechanical threshold was implemented using a digital analgesimeter through increasing pressure method. The nociceptive mechanical threshold was defined as the minimum force necessary to trigger the paw withdrawal reflex. Withdrawal threshold was assessed through application of pressure ranging from 0 to 50 g with an accuracy of 0.2 g. Voluntary movements associated with locomotion were not considered a withdrawal response. Stimuli were applied at 5 s intervals. The measurements were repeated 3 times and the final value was obtained by average of the 3 measurements.

2.4. Euthanasia and Sample Collection

The animals were euthanized on the 14th day, after the mechanical nociceptive threshold evaluation, through a propofol overdose (150 mg/kg). The spinal cord was collected through hydraulic extrusion technique [23]. The lumbosacral region of the spinal cord (9 mm) was removed and segmented into three fragments (3 mm) for storage and subsequent analysis. Two fragments were stored in RNAse- and DNAse-free cryovials at −80 °C for subsequent enzymatic and PCR analysis. The third fragment was stored in 4% paraformaldehyde for 24 h with subsequent change to 70% alcohol, followed by tissue preparation, changing solutions to 80% alcohol (50 min), 90% alcohol (50 min), absolute alcohol (50 min), xylene (50 min) and paraffin (30 min, controlled temperature between 55 °C and 60 °C), with ensuing embedding in histological paraffin at the same temperature. Then, histological sections with a thickness of 4 µm were obtained and placed on previously gelatinized slides.

2.5. Enzyme Analyses

2.5.1. Catalase Activity Analysis

The analysis of catalase activity (CAT) was carried out using a spectrophotometer device (Thermo Scientific GENESYS 6™ UV-Vis, Waltham, MA, USA) with the wavelength set at 240 nM, with readings through quartz cuvettes. For the “blank” solution, 50 mM potassium phosphate buffer was used with pH adjusted to 7, and the device was calibrated to zero. Soon after, 40 µL of a hydrogen peroxide (H2O2) solution was added, prepared using 200 µL of H2O2 and 9.8 mL of distilled water, protected from light. The reading was then collected every 15 s for 45 s; then, 9 µL of the sample was added and the reading was carried out until 150 s.

2.5.2. Analysis of Superoxide Dismutase Activity

The analysis of Superoxide Dismutase (SOD) activity was also executed using a spectrophotometer (Thermo Scientific GENESYS 6™ UV-Vis, Waltham, MA, USA), using a wavelength of 420 nM, with readings through quartz cuvettes. The “blank” solution was prepared by addition of 1200 µL of TFK and 32 µL of EDTA, and the device was calibrated to zero. Soon after, 154 µL of the pyrogallol solution was added and the absorbance was read for 5 min every 30 s, to obtain the absorbance values of the pyrogallol auto-oxidation. At 5 min of the reaction, 154 µL of the sample was added to the cuvette, and readings continued every 30 s during 10 min of reaction.

2.6. Protein Quantification

The protein concentration of the sample was quantified using the Bradford method (1976), based on the principle of binding of the dye to the protein. The method uses bovine serum protein (BSA), calculating the protein concentration of the sample using a standard curve of BSA dilution at 0.1 mg/mL. Amounts of 150 μL of BSA at a concentration of 0.1 mg/mL and 50 μL of TFK (50 mM, pH 7.0) were placed on a plate. An amount of 10 μL of the sample was diluted in 40 μL of TFK (1:20) and then added in triplicate. An amount of 200 μL of Coomassie Blue was applied under the plate in all wells (Sigma B-0770, Sigma Aldrich Corporation, St. Louis, MO, USA). The absorbance was then read at a wavelength of 595 nm on the spectrophotometer device (SpectraMax Paradigm–Multi-Mode Detection Platform, Molecular Devices, LLC, San Jose, CA, USA).

2.7. Immunohistochemistry

For immunohistochemical evaluation, the streptavidin–biotin–peroxidase technique was used, using the EnVisionTM FLEX+, Mouse, High pH kit (Agilent Dako®, Santa Clara, CA, USA). Five histological sections from each animal were analyzed for each antibody. The protocol followed that recommended by the kit specifications. A prior standardization process appropriate to the tissue defined the time in each solution. The histological sections on the slides were heated in an oven, deparaffinized in xylene PA, rehydrated in absolute ethyl alcohol (100%, 90%, 80% and 70%, respectively), washed and immersed in citrate buffer solution (citric acid), pH 6, in a water bath at 90–98 °C for 20 min, and subsequently remained in citrate buffer for another 20 min at room temperature, followed by wash buffer washing solution (DM831, Dako®, Santa Clara, CA, USA).
Then, treatment started with a sequence based on Illie M. et al. (2017) [24], initially with peroxidase blocking (SM801, Dako®, Santa Clara, CA, USA), serum blocking (protein block) and incubation in a humidity chamber for 30 min at room temperature. The sections were soaked in the primary antibody of choice and were incubated overnight for 18 h in a humidity chamber under refrigeration at 4 °C. The utilized antibodies (Santa Cruz Biotechnology®, Paso Robles, CA, USA), as well as species, dilution, code and DAB development time, are described in Table 1.
The process continued the next day by washing solution, then by addition of stabilizing protein (SM804, Dako®, Santa Clara, CA, USA) for 30 min in a humidity chamber at room temperature, followed by washing one more time. The secondary antibody (SM802, Dako®, Santa Clara, CA, USA) was applied on the slides for 30 min and thereupon washed. The cromogen used was diaminobenzidine (DAB) code DM827 (Dako®, Santa Clara, CA, USA), in a dilution of 1:50, protected from light, with a time defined by standardization through appropriate labeling tests for each antibody. The slides were then stained in hematoxylin (HE), followed by dehydration in ethyl alcohol (70%, 80%, 90% and 100%, respectively) and xylene. Later, the slides were mounted using Canada balsam and coverslips, for later reading under an optical microscope.

2.8. Quantitative PCR

qRT-PCR technique was performed. Initial extraction of total RNA from the spinal cord was performed using Trizol, following the manufacturer’s instructions (Invitrogen, Life Technologies, Carlsbad, CA, USA). The method consisted of an initial stage of tissue lysis and homogenization for 5 min at room temperature to completely dissociate the nucleoprotein complexes. The lysate was transferred to a 1.5 mL microtube, and 0.2 mL of chloroform was added, followed by 15 s of homogenization, 2 to 3 min of incubation at room temperature and centrifugation at 12,000× g for 15 min at 4 °C for separation into three phases; RNA encompassed the colorless phase. In the third stage, the colorless phase was transferred to a new tube, with addition of 0.5 mL of isopropyl alcohol and incubation for 30 min at −80 °C, followed by centrifugation at 12,000× g for 10 min at 4 °C to precipitate the RNA. The supernatant was discarded and the pellet was placed on ice. Then, the pellets were washed with 1 mL of 75% ethanol, homogenized and centrifuged at 10,500× g for 5 min at 4 °C. After discarding the ethanol, the RNA pellet was solubilized in RNAse- and DNAse-free water and immediately stored at −80 °C. Analysis of the concentration and quality of RNA in the tissue of each sample was carried out using a nanodrop 2000 Spectrophotometer (Thermo Scientific, Waltham, MA, USA).
The commercial kit GoTaq® qPCR 1 µg of RNA was used for reverse transcription reactions using RT-qPCR Systems (A6010, PROMEGA, Promega Corporation, Madison, WI, USA). The quantification of the target gene transcripts was conducted through qPCR using SYBR Green on the Applied Biosystems® 7500 Real-Time PCR System equipment (Applied Biosystems, Foster City, CA, USA). The reactions were performed using 1.5 μL of cDNA, 100 nM of each primer and 12.5 μL of the GoTaq® qPCR Master Mix reagent, 2X, in a final volume of 20 μL of reaction. The DNA amplification mix was used as a negative control and the cDNA sample replaced by water. Primers for Grp78, Chop, Ho1, Perk, Bax, Bcl-xL, Casp 3, Casp 9, Il-6, Il-10, Il-18 and Il-1 β were designed based on the Rattus norvegicus mRNA sequence (Table 2). The calculations of the gene expression were performed using the 2−ΔΔCT method, and the results obtained for each group were compared quantitatively after normalization based on the expression of Gadph Rattus norvegicus.

2.9. Statistical Analyses

Statistical analysis was performed using Graph Pad Prism 5.01 software. A completely randomized design was used. Nociceptive threshold data were analyzed using two-way analysis of variance (two-way ANOVA) followed by the Bonferroni test, and the results were expressed as mean ± standard error of the mean. For immunohistochemical analysis, Student–Newman–Keuls (SNK) test was used for multiple comparisons. The ID50 of the doses used in the experiment was calculated from a dose–response curve using nonlinear regression. A significance level of 5% was adopted in all analyses.

3. Results

3.1. Effect of Chemotherapy on Mechanical Pain Hypersensitivity

Daily administration of intraperitoneal vincristine (0.05 mg/kg) was capable of inducing mechanical hyperalgesia, evidenced by a decrease in the mechanical nociceptive threshold. Neuropathy could be observed from the fourth day (Figure 1) with a low threshold maintained in the positive control until the 14th day of the experiment, with a cumulative effect. The naïve (29.49 ± 0.19 g) and negative (29.66 ± 0.17 g) groups differed from the positive control group (14.50 ± 0.93 g), demonstrating that neuropathic pain was established through the chemotherapy drug, vincristine.

3.2. Amantadine Increased the Nociceptive Mechanical Threshold in Rats with Vincristine-Induced Peripheral Neuropathy

The administration of amantadine had a positive effect on preventing the development of mechanical hyperalgesia at 25 mg/kg (23.09 ± 0.62 g) and 50 mg/kg, showing a significant difference from the positive control group (24.69 ± 0.56 g). The ID50 (calculated from the effective dose capable of increasing the nociceptive mechanical threshold by 50%) was 9.58 mg/kg. The doses of 2 mg, 5 mg and 12 mg did not show a significant difference (p > 0.05) compared to the positive control group (14.50 ± 0.95 g) (Figure 1).

3.3. Amantadine Treatment Inhibits the Expression of Pro-Inflammatory Cytokines in the Spinal Cord of Rats with Vincristine-Induced Peripheral Neuropathy

Pro-inflammatory cytokine expressions of IL-6, TNF-α and MIP-1α, tested through an immunohistochemical technique, demonstrated a statistically significant increase (p = 0.0001) in groups that received a daily intraperitoneal injection of vincristine, evidencing the increase in these cytokines in the inflammatory process of chemotherapy-induced neuropathy (Figure 2). Amantadine was able to significantly reduce (p < 0.0001) the expression of Il-6 in the spinal cord of rats with chemotherapy-induced neuropathy, which is demonstrated by the statistical difference in relation to the positive control group; its highest effect was observed at the dose of 50 mg/kg. Similarly, the TNF-α expression was reduced in groups treated with amantadine; the 50 mg/kg group showed lower immunostaining (Figure 2).
MIP-1α also showed a decrease (p < 0.0001) in its expression in animals treated with amantadine, with the reduction being more pronounced at the highest dose tested (50 mg/kg), suggesting a role for the macrophage inflammatory protein in the mechanism of chemotherapy neuropathy and the anti-inflammatory mechanism of amantadine. The same occurred with CX3CR1, demonstrating a dose-dependent decrease (p < 0.0001). In the immunostaining of the anti-inflammatory cytokine interleukin-10, an increase was identified in animals that received amantadine (Figure 3). The expression of CXCR4 immunostaining did not demonstrate a statistically significant difference between the groups (Figure 3).
SOD and CAT immunostaining expression was significantly increased in animals that received amantadine at 50 mg/kg (p = 0.0001), which evidences the role of amantadine in oxidative stress. The dose of 25 mg/kg did not demonstrate significant expression of SOD and CAT, suggesting an antioxidant effect only at the highest tested dose. On the other hand, GPx immunostaining did not exhibit a significant difference between the positive control group and the group that received amantadine; it was only greater in the groups that received vincristine in relation to the control groups without treatment and saline solution, suggesting an induction of antioxidant release to oppose chemotherapy-induced neuropathy.

3.4. Amantadine Treatment Increased the Expression of Antioxidant Enzymes (CAT and SOD) in the Spinal Cord of Rats with Vincristine-Induced Peripheral Neuropathy

Vincristine played an important role in the statistically significant decrease (p < 0.0001) in the antioxidant enzymes catalase (CAT) and Superoxide Dismutase (SOD) (p < 0.0001), which is demonstrated by the difference between the positive control group in relation to the naïve and saline solution groups (Figure 4). Treatment with amantadine proved to be effective in a dose-dependent manner, once it increased SOD and CAT (p = 0.0001) activity, corroborating the results obtained with immunohistochemistry (Figure 4).

3.5. Amantadine Treatment Inhibits the Expression of Reticular Stress Mediators in the Spinal Cord of Rats with Vincristine-Induced Peripheral Neuropathy

Vincristine-induced peripheral neuropathy in rats can cause endoplasmic reticulum stress in the spinal cord. Quantitative PCR (qPCR) was performed to verify whether treatment with amantadine at the doses used could prevent the occurrence of such. For this purpose, misfolded-protein activation-pathway key mediators, that indicate the occurrence of reticular stress, were analyzed, Grp78, CHOP, Perk and Ho1.
Grp78 gene expression showed no significant difference between groups (Figure 5). As for CHOP expression, the 25 and 50 mg/kg treatments with amantadine induced significantly lesser values for changes caused by neuropathy, when compared to the positive control group (Figure 5, p < 0.01).
Peripheral neuropathy increased Perk gene expression (Figure 5, p < 0.01), compared to the negative control. Amantadine 50 mg/kg was the only dose to cause a significant reduction in its gene expression. Concerning reticular stress signal (Ho1) expression, treatments with amantadine 25 and 50 mg/kg significantly reduced its expression caused by neuropathy, compared to the positive control group (Figure 5; p < 0.001). Altogether, these data show that peripheral neuropathy up-regulates the expression of genes related to UPR and reticular stress in the spinal cord of rats; also, amantadine moderates their expression.
Since Bcl-x is the predominant protein of the Bcl-2 family and one of the main regulators of apoptosis, and the Bcl-xL/Bax protein ratio is a good predictor of cell survival, we also evaluated the gene expression of both markers. Peripheral neuropathy induced by vincristine (positive control group) down-regulates Bcl-xl expression, while up-regulating Bax gene expression, with a significant difference in the group treated with amantadine 50 mg/kg (Figure 6; p < 0.05).
Regarding the expression of CASP 3 and CASP 9, groups treated with amantadine 25 mg and 50 mg/kg significantly reduced the spinal cord gene expression caused by neuropathy (Figure 6; p < 0.05). Accordingly, these data show that peripheral neuropathy promotes the expression of immunological and apoptotic markers and that treatment with amantadine was able to therapeutically modulate this response.

4. Discussion

4.1. Vincristine Induces Neuropathic Pain in Chemotherapy

In the present study, we demonstrated that daily intraperitoneal administration of vincristine (50 µg/kg) was able to induce mechanical hyperalgesia in rats. The dose of vincristine used in this study was based on a previous study by Nozaki-Taguchi et al., 2001 [22], who established a chemotherapy-induced neuropathy model with administration of intravenous vincristine 30–100 µg/kg for 14 days. The first measurement of the mechanical threshold occurred on the seventh day, which revealed tactile allodynia observed in doses greater than 50 µg/kg/day, in a dose-dependent manner.
The hyperalgesia observed in the present study was also perceived in other studies conducted by Authier et al., 2003 [25], where they demonstrated sustained allodynia and mechanical hyperalgesia in rats when doses of vincristine were administered for 10 days (50, 100 and 150 μg/kg IV), by Kiguchi et al. (2008) [26], with doses of 10–100 μg/kg, by Chiba et al., 2017) [27], with doses of 100 µg/kg/day for 7 and 14 days, and by Vashistha, Sharma and Jain, 2016 [28], where they observed a reduction in the mechanical nociceptive threshold in rats with vincristine (50 ug/kg/day, IP) for 11 days.
Until the day of the present study, there were insufficient data regarding the combination of amantadine and vincristine in the same medical protocol to assess potential risks, such as side effects, the decrease in efficacy of the anti-cancer treatment, and the development of viral resistance. A study in 2020 aimed to assess the use of amantadine to evaluate the chemotherapy response in lung cancer. Systemic therapy choices were varied, including a combination of chemotherapeutic drugs such as cisplatin, carboplatin, pemetrexed and others. No toxicities or side effects were observed with the addition of amantadine to chemotherapy. However, the study did not mention the use of vincristine [29]. Nevertheless, further research is recommended to investigate potential interactions and risks associated with their concurrent use.

4.2. Amantadine Alleviates Mechanical Hyperalgesia in Chemotherapy-Induced Neuropathic Pain

By antagonizing NMDA receptors, modulating glutamatergic neurotransmission and inhibiting central sensitization processes that contribute to chronic pain, NMDA antagonist drugs may normalize neuronal activity and reduce the hyperexcitability associated with neuropathic pain [19].
Some of the primary medications utilized for pain management during cancer chemotherapy include NMDA antagonists like ketamine and methadone. Both drugs have extensive research supporting their efficacy in managing neuropathic pain, with ketamine noted for its rapid pain relief onset and effectiveness across acute and chronic conditions [30]. However, ketamine also presents risks such as psychotomimetic effects and potential for abuse, necessitating vigilant monitoring [30]. Methadone, on the other hand, offers prolonged pain relief with lower respiratory depression risks compared to other opioids, though it requires careful monitoring for drug interactions and cardiac side effects like QT interval prolongation [31].
Amantadine and memantine, while showing promise as potential treatments for neuropathic pain, lack robust evidence compared to ketamine and methadone. Both drugs are generally well tolerated, with minimal sedative effects and potential neuroprotective properties [32,33]. Memantine has demonstrated cognitive enhancement and neuroprotection in specific neurological conditions, but may cause gastrointestinal issues like nausea and diarrhea, necessitating careful dosing and monitoring [16]. Amantadine, known for its neuroprotective effects, has been used successfully in combination therapies but can lead to CNS side effects like nausea, orthostatic hypotension, dizziness and confusion, requiring adjustments in patients with renal impairment. Further research is essential to delineate their specific effects on neuropathic pain patients [34,35].
The doses used in the present study were based on previous studies in rats whose effective doses ranged from 10 mg [36] to 135 mg [37]. In our study, it was demonstrated that doses of 2, 5 and 12 mg/kg were not effective in improving mechanical hyperalgesia in chemotherapy-induced neuropathy in rats. The effective doses improving mechanical hyperalgesia were 25 and 50 mg/kg, indicating a statistical difference from the positive control group. The dosage commonly used by humans, particularly in Parkinson’s disease patients where its use is well documented, generally tolerates doses up to 200 mg of conventional amantadine. This dose range is widely considered safe and well tolerated [38]. Muller et al., in 2022 [38], demonstrated that extended-release amantadine capsules provide enhanced convenience with a steady and sustained release of amantadine over time, leading to improved tolerability and efficacy compared to traditional immediate-release formulations. The use of dosages such as 25 mg/kg/day and 50 mg/kg/day in patients with neuropathic pain remains to be elucidated regarding safety and side effects.
Previous studies with amantadine demonstrated a positive effect in different models of neuropathic pain, on both animals and humans. In a study in dogs with refractory osteoarthritis, the use of 3–5 mg/kg demonstrated effective improvements in chronic pain, associated with an NSAID (meloxicam) [39]. Previous denervation of the sciatic nerve study in rats [40] showed that 90 mg/kg promoted a reduction in nociception in a model of neuropathic pain. The drug was also effective in postoperative cognitive dysfunction in rats at a dose of 25 mg/kg/day [41]. In a study with a traumatic brain injury (TBI) model in rats, doses of 45 and 135 mg/kg were able to reduce post-TBI depression behavior [30]. In humans, amantadine effectively reduced pathological low-back pain in patients with neuropathic pain [32], surgical neuropathic pain in cancer patients [33] and peripheral sensitization in patients with chronic low-back pain [42]. Pud and collaborators, in 1998, concluded that, in models in which NMDA receptor activation-dependent neuropathic pain is possible, amantadine can effectively reduce the existing neuropathic pain [33].
The findings from our study also indicate that the antinociceptive effect of amantadine slightly decreased within the 14-day study period, raising questions about its long-term efficacy. Fumagalli et al., 2021, suggested that neuronal damage might precede the inflammatory process in various preclinical models of chemotherapy-induced peripheral neuropathy, potentially leading to neuropathy progression over time [43]. This progression could involve increased inflammation or neuronal injury that may not be effectively managed solely by NMDA antagonists.

4.3. Regulation of Inflammatory Mediators by Amantadine Demonstrates Anti-Inflammatory Effects in Chemotherapy-Induced Neuropathy

Vincristine was able to induce an inflammatory response in the spinal cord, evidenced by an increase in immunostaining through an immunohistochemical technique in the positive control group of the pro-inflammatory cytokines IL-6, TNF-α, MIP-1α, IL-18 and IL-1 β. IL-6 has been shown to be an inflammatory cytokine with a fundamental role in pathological pain, acting on CIPN [44]. It is directly linked to nociceptive plasticity, increasing transduction in sensory neurons [45], and thus contributes to both nociceptor sensitization and central sensitization [46,47,48].
TNF-α is of great importance in pro-inflammatory processes. It is considered the main mediator in processes that involve necrosis, apoptosis or proliferation, being a suitable systemic marker of tissue damage [49,50], regulating several types of glutamatergic receptors [51,52], and is involved in the production of the neurotransmitter glutamate, as well as decreasing glutamate reuptake, raising it to toxic levels [53]. In the dorsal horn of the spinal cord, TNF-α increases glutamate release from TRPV1-expressing C-fiber terminals, leading to increased excitatory synaptic transmission on interneurons in lamina II, which in turn create synapses on lamina I projection neurons to form a pain circuit [54]. The reduction in TNF-α immunostaining in animals treated with amantadine suggests the inhibition of this circuit.
There was a decreased expression of MIP-1α, in animals treated with 25 and 50 mg/kg of amantadine in dorsal medulla neurons. Macrophage inflammatory protein is an inflammatory chemokine that sensitizes TRPV1, which is sensitive to noxious stimuli receptors and is an important mediator in pathological pain, through its receptor CCR1, suggesting that this chemokine may participate in neuropathic pain through glial cell crosstalk [55]. Likewise, in a model of neuropathy induced by sciatic nerve ligation, an increase in MIP-1α was observed in animals with CIPN [56].
Amantadine’s ability to regulate inflammatory mediators contributes to its neuroprotective effects. By mitigating the inflammatory response, it prevents further neuronal damage and promotes the survival and function of neurons, which is critical in managing chronic pain conditions like CIPN [14,15,16,17,18,19]. In the present study, the administration of amantadine 25 and 50 mg/kg proved to effectively reduce pro-inflammatory cytokine expression (IL-6, TNF-α, MIP-1α, IL-18 and IL-1β), and increase cytokine anti-inflammatory IL-10 expression in the spinal cord of rats treated daily. This proposes an anti-inflammatory effect of amantadine in CIPN. Similarly, in a rat spinal cord injury model, amantadine exhibited a protective effect with reduced neuroinflammation and oxidative stress [57] in post-operative rats [33], and in a sepsis model induced by cecal ligation and puncture [58]. Milligan et al., 2006 [59] confirmed that the pharmacological application of Il-10 had positive results attenuating hyperalgesia and allodynia behaviors across various pain models, demonstrating its effectiveness in alleviating pain symptoms.
Our results demonstrate that CX3CR1 expression was up-regulated in the spinal cord of vincristine-treated animals, whereas it was down-regulated by amantadine treatment, in a dose-dependent manner. The same occurred with a study using mitokine in the chemotherapy neuropathy model with vincristine [60]. CX3CR1 is expressed in the dorsal horn of the spinal cord and mediates neuron–glia communication; in the case of peripheral neuropathy with interruption of homeostasis, it increases signaling in the dorsal horn of the spinal cord, thus increasing maladaptive neuron–glia signaling. This contributes importantly to the amplification of nociceptive transmission during the neuropathy process [61].
The alpha-chemokine receptor (CXCR4) participates in the development and maintenance of neuropathic pain mediating the transport of progenitor cells in the bone marrow [62], having also demonstrated the modulation of neuropathic pain through the activation of the ERK pathway [63]. Our results demonstrate that there was no statistically significant difference between the groups. In the same way, Deng et al., 2012 [64], in a chemotherapy-induced neuropathy model with cisplatin and paclitaxel, concluded that CXCR4 signaling did not contribute to the maintenance of chemotherapy-induced neuropathy, and that blocking CXCR4 signaling with AMD3100 failed to reverse the established chemotherapy-induced neuropathy [65].
The neuroinflammatory modulation induced by amantadine at doses of 25 and 50 mg/kg in this study points to an important therapeutic target in the management of CIPN, in view of the fact that, with these glial mediators (IL-6, TNF-α, MIP-1α and IL-10), CX3CR1 can pre-eminently modulate excitatory and inhibitory synaptic transmission, leading to sensitization and enhanced chronic pain states.

4.4. The Treatment with Amantadine Was Effective in Increasing Antioxidant Enzymes in the Cord of Rats with Vincristine-Induced Peripheral Neuropathy

Vincristine significantly decreased antioxidant enzymes catalase (CAT) and Superoxide Dismutase (SOD). This is in agreement with previous studies that identified a relationship between CIPN induced by vincristine and oxidative stress [66,67,68,69].
Damage to peripheral nerves may involve oxidative stress mediated by mitochondria, damage to the myelin sheath and other antioxidant enzymes, and the explanation of the mechanisms by which the oxidative stress pathway acts in neuropathic pain may be useful to increase the chances of therapeutic protocol improvement for CIPN patients [70]. SOD participates in the process of neuropathic pain, since the superoxide anion radical, produced via xanthine oxidase, and nitric oxide, as a precursor of peroxynitrite in NMDA, are involved in central sensitization mediation [71].
Treatment with amantadine was effective in increasing SOD and CAT. In accordance with our findings, Mata-Bermudez et al., in 2021 [20], demonstrated the role of amantadine in oxidative stress in a model of spinal cord injury in rats. Orhan et al., in 2021 [72], obtained a similar result under oxidative stress in a model of ischemia and reperfusion injury in the hind limb of rats, using amantadine. In a model of traumatic spinal cord injury in rats, Dogan and Karaka in 2020 [57] had similar effects with the administration of amantadine, inhibiting oxidative stress and leading to improvement in neuropathic pain.
Dogan and Karaka, 2020 [57] and Orhan et al. 2021 [72] observed an increase in GPx expression in models of spinal cord injury and ischemia and reperfusion injury in the hind limb of rats, respectively. In the present study, this finding could not be identified, as the treated groups did not obtain a significant statistical difference between them, which demonstrates that the amantadine antioxidant pathway releases antioxidant enzymes in chemotherapy-induced neuropathy, with a significant increase in SOD and CAT in the tested groups, but not GPx, thus encouraging further research based on this pathway for onward elucidation. Although there was no significant difference between the groups, there was a trend towards increased antioxidant activities. Other factors may have influenced the quantification of sample activity, such as the presence of endogenous inhibitors or activators (Figure 4).

4.5. Amantadine Treatment Inhibits the Expression of Reticular Stress Mediators in the Spinal Cord of Rats with Vincristine-Induced Peripheral Neuropathy

In this peripheral neuropathy model, vincristine was found to contribute to inducing endoplasmic reticulum stress, as evidenced by the increased gene expression of CHOP, PerK and Ho1 in the quantitative analysis of PCR, as observed in other studies of neuropathic pain-induced reticulum endoplasmic stress in the rat spinal cord [73]. Treatment with amantadine at doses of 25 mg/kg and 50 mg/kg was able to reduce the gene expression of CHOP and Ho1, and only the dose of 50 mg/kg was able to reduce PerK. Similarly, a study that sought to demonstrate the effectiveness of ketamine, another NMDA antagonist, on endoplasmic reticulum stress in the process of neuropathic pain concluded that the drug was effective in reducing ER stress markers, suggesting that there may be a relationship with the NMDA pathway [74].

4.6. Modulatory Effect of the Spinal Cord Response to the Apoptotic Mechanisms of Amantadine in Chemotherapy-Induced Neuropathy

The process of apoptosis is one of the main pathways for neuron death during spinal cord injury [75]. In this process, caspases, and proteins from the Bcl-2 family, such as Bcl-x (pro-survival protein), play a major role in determining cell death or survival. Considering that Bcl-x is the predominant protein of the Bcl-2 family, one of the main regulators of apoptosis, and that the Bcl-xL/Bax protein ratio is a good predictor of cell survival, we also evaluated the gene expression of these markers. Vincristine-induced peripheral neuropathy (positive control group) reduced Bcl-xl expression and increased Bax gene expression, which corroborates studies of apoptosis in spinal cord injury [76]. The expressions of CASP 3 and CASP 9 were also higher in the untreated neuropathy group, while there was a significant decrease in the expression of these caspases in spinal cord tissue affected by vincristine after treatment with amantadine. Similarly, in a study involving glutamate [77], the use of amantadine led to a decrease in the apoptotic index. This indicates its potential in minimizing cell death in the cerebral cortex.

5. Conclusions

Amantadine demonstrated efficacy in reducing mechanical hyperalgesia in animals treated with 25 and 50 mg/kg/day, in a dose-dependent manner. It presented an anti-inflammatory effect through the activation of anti-inflammatory cytokines and decreased expression of pro-inflammatory cytokines. An antioxidant effect was demonstrated through the increased expression of the antioxidant enzymes SOD and CAT, and regulation of apoptotic mediators. Our data indicate that amantadine may be a potential alternative for the treatment of vincristine-induced peripheral neuropathic pain, although the study suggests that further investigation is needed regarding drug interactions, dosing, effectiveness in humans and potential side effects.

Author Contributions

M.S.L.d.L. conceived and designed the experiments. The experiments and data collection were performed by I.S.A.D., J.N.S.d.O., R.V.N., Á.J.C.S., I.S.d.O., S.S.d.S. and C.S.d.C.M. Data were analyzed by M.S.L.d.L. and J.F.S. I.S.A.D. wrote the article. I.S.A.D. and R.V.N. analyzed and arranged data for statistical analysis. I.S.A.D. and J.M.X.C. performed the statistical analyses. All authors have read and agreed to the published version of the manuscript.

Funding

This project was financed by CNPQ (Grant number 423044/2018-4); R.V.N. (CNPQ 140816/2020-8); (CAPES, number: 88881.693767/2022-01). This work also received support from FAPESB: I.S.A.D. (0144/2020) and State University of Santa Cruz (UESC) (number: 073.6762.2024.0018126-27).

Institutional Review Board Statement

The animal study protocol was approved by the Ethics Committee of Use of Animals (CEUA) of State University of Santa Cruz–UESC (protocol 033/18, 13 March 2019), and followed the guidelines on the care and use of laboratory animals issued by the National Council for Animal Experimentation Control (Brazil).

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Acknowledgments

The authors would like to express sincere gratitude to Marcia Silva and Ivo Arouca for their assistance and technical support, to Jeane Cordeiro and Luciano Cardoso for their support and shared knowledge, and to Carlos Priminho Pirovani for the shared expertise.

Conflicts of Interest

The authors declare no conflicts of interest. The founders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Bray, F.; Laversanne, M.; Cao, B.; Varghese, C.; Mikkelsen, B.; Weiderpass, E.; Soerjomataram, I. Comparing cancer and cardiovascular disease trends in 20 middle- or high-income countries 2000–2019: A pointer to national trajectories towards achieving Sustainable Development goal target. Cancer Treat. Rev. 2021, 100, 102290. [Google Scholar] [CrossRef] [PubMed]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  3. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Mathers, C.; Parkin, D.; Piñeros, M.; Bray, A. Estimating the global cancer incidence and mortality in 2018: Globocan sources and methods. Int. J. Cancer 2020, 144, 1941–1953. [Google Scholar] [CrossRef] [PubMed]
  4. World Health Organization (WHO). Global Health Estimates 2020: Deaths by Cause, Age, Sex, by Country and by Region, 2000–2019; WHO: Geneva, Switzerland, 2020; Available online: https://who.int/data/gho/data/themes/mortality-and-global-health-estimates/ghe-leading-causes-of-death (accessed on 4 April 2022).
  5. Cavaletti, G.; Alberti, P.; Marmiroli, P. Chemotherapy-induced peripheral neurotoxicity in the era of pharmacogenomics. Lancet Oncol. 2017, 12, 1151–1161. [Google Scholar] [CrossRef] [PubMed]
  6. Noble, C.O.; Guo, Z.; Hayes, M.E.; Marks, J.D.; Park, J.W.; Benz, C.C.; Drummond, D.C. Characterization of highly stable liposomal and immunoliposomal formulations of vincristine and vinblastine. Cancer Chemother. Pharmacol. 2009, 64, 741–751. [Google Scholar] [CrossRef] [PubMed]
  7. Peters, C.M.; Jimenez-Andrade, J.M.; Jonas, B.M.; Sevcik, M.A.; Koewler, N.J.; Ghilardi, J.R.; Wong, G.Y.; Mantyh, P.W. Intravenous paclitaxel administration in the rat induces a peripheral sensory neuropathy characterized by macrophage infiltration and injury to sensory neurons and their supporting cells. Exp. Neurol. 2007, 203, 42–54. [Google Scholar] [CrossRef] [PubMed]
  8. Starobova, H.; Vetter, I. Pathophysiology of Chemotherapy-Induced Peripheral Neuropathy. Front. Mol. Neurosci. 2017, 10, 174–195. [Google Scholar] [CrossRef] [PubMed]
  9. Ma, J.; Kavelaars, A.; Dougherty, P.M.; Heijnen, C.J. Beyond symptomatic relief for chemotherapy-induced peripheral neuropathy: Targeting the source. Cancer 2018, 124, 2289–2298. [Google Scholar] [CrossRef] [PubMed]
  10. Marmiroli, P.; Scuteria, A.; Cornblath, D.R.; Cavaletti, G. Pain in chemotherapy-induced peripheral neurotoxicity. J. Peripher. Nerv. Syst. 2017, 22, 156–161. [Google Scholar] [CrossRef] [PubMed]
  11. Boehmerle, W.; Huehnchen, P.; Peruzzaro, S.; Balkaya, M.; Endres, M. Electrophysiological, behavioral and histological characterization of paclitaxel, cisplatin, vincristine and bortezomib-induced neuropathy in C57Bl/6 mice. Sci. Rep. 2014, 4, 6370. [Google Scholar] [CrossRef] [PubMed]
  12. Seretny, M.; Currie, G.L.; Sena, E.S.; Ramnarine, S.; Grant, R.; MacLeod, M.R.; Colvin, L.A.; Fallon, M. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: A systematic review and meta-analysis. Pain 2014, 155, 2461–2470. [Google Scholar] [CrossRef] [PubMed]
  13. Gaynor, J.S.; Muir, I.W. Manual of Pain Control in Veterinary Medicine, 2nd ed.; Mosby: St. Louis, MO, USA, 2009; Volume 2, pp. 13–41. [Google Scholar]
  14. Childers, W.E.; Baudy, R.B. N-Methyl-d-Aspartate Antagonists and Neuropathic Pain: The Search for Relief. J. Med. Chem. 2007, 50, 2557–2562. [Google Scholar] [CrossRef] [PubMed]
  15. Bozic, M.; Valdivielso, J.M. The potential of targeting NMDA receptors outside the CNS. Expert Opin. Ther. Targets 2015, 19, 399–413. [Google Scholar] [CrossRef] [PubMed]
  16. Pickering, G.; Morel, V. Memantine for the treatment of general neuropathic pain: A narrative review. Fundam. Clin. Pharmacol. 2017, 32, 4–13. [Google Scholar] [CrossRef] [PubMed]
  17. Blanpied, T.A.; Clarke, R.J.; Johnson, J.W. Amantadines inhibits NMDA receptors by accelerating channel closure during channel block. J. Neurosci. 2005, 25, 312. [Google Scholar] [CrossRef] [PubMed]
  18. Schoen, B.; Eickmeyer, S. Poster 52 Acute Hallucinosis Related to Amantadine Use in the Setting of Traumatic Brain Injury: A Case Report. PM&R J. Inj. Funct. Rehabil. 2016, 8, 178. [Google Scholar] [CrossRef] [PubMed]
  19. Aiyer, R.; Mehta, N.; Gungor, S.; Gulati, A. Systematic Review of NMDA Receptor Antagonists for Treatment of Neuropathic Pain in Clinical Practice. Clin. J. Pain 2016, 34, 450–467. [Google Scholar] [CrossRef] [PubMed]
  20. Mata-Bermudez, A.; Rios, C.; Burelo, M.; Pérez-González, C.; García-Martínez, B.A.; Jardon-Guadarrama, G.; Calderón-Estrella, F.; Manning-Balpuesta, N.; Diaz-Ruiz, A. Amantadine prevented hypersensitivity and decreased oxidative stress by NMDA receptor antagonism after spinal cord injury in rats. Eur. J. Pain 2021, 25, 1839–1851. [Google Scholar] [CrossRef] [PubMed]
  21. Kerschner, D.; Von Ritgen, S. Amantadin, ein NMDA-Rezeptorantagonist, zur Behandlung von neuropathischen Schmerzen bei einer Katze. Vet. Med. Austria 2021, 108, 205–212. [Google Scholar]
  22. Nozaki-Taguchi, N.; Chaplan, S.R.; Higuera, E.S.; Ajakwe, R.C.; Yaksh, T.L. Vincristine-induced allodynia in the rat. Pain 2001, 93, 69–76. [Google Scholar] [CrossRef] [PubMed]
  23. Moghaddasi, R. An ex vivo preparation of mature mice spinal Cord to study synaptic transmission on motoneurons. J. Neurosci. Methods 2007, 159, 1–7. [Google Scholar] [CrossRef]
  24. Ilie, M.; Khambata-Ford, S.; Copie-Bergman, C.; Huang, L.; Juco, J.; Hofman, V. Use of the 22C3 anti–PD-L1 antibody to determine PD-L1 expression in multiple automated immunohistochemistry platforms. PLoS ONE 2017, 12, e0183023. [Google Scholar] [CrossRef] [PubMed]
  25. Authier, N.; Gillet, J.P.; Fialip, J.; Eschalier, A.; Coudore, F.A. New Animal Model of Vincristine-Induced Nociceptive Peripheral Neuropathy. NeuroToxicology 2003, 24, 797–805. [Google Scholar] [CrossRef] [PubMed]
  26. Kiguchi, N.; Maeda, T.; Kobayashi, Y.; Kondo, T.; Ozaki, M.; Kishioka, S. The critical role of invading peripheral macrophage-derived interleukin-6 in vincristine-induced mechanical allodynia in mice. Eur. J. Pharmacol. 2008, 592, 87–92. [Google Scholar] [CrossRef] [PubMed]
  27. Chiba, T.; Okay, Y.; Sashida, H.; Kanbe, T.; Abe, K.; Utsunomiya, I.; Taguchi, K. Vincristine-induced peripheral neuropathic pain and expression of transient receptor potential vanilloid 1 in rat. J. Pharmacol. Sci. 2017, 133, 254–260. [Google Scholar] [CrossRef] [PubMed]
  28. Vashistha, B.; Sharma, A.; Jain, V. Ameliorative potential of ferulic acid in vincristine-induced painful neuropathy in rats: An evidence of behavioral and biochemical examination. Nutr. Neurosci. 2016, 20, 60–70. [Google Scholar] [CrossRef] [PubMed]
  29. Maksymiuk, A.W.; Tappia, P.S.; Bux, R.A.; Moyer, D.; Huang, G.; Joubert, P.; Sitar, D.S. Use of Amantadine in the Evaluation of Response to Chemotherapy in Lung Cancer: A Pilot Study. Futur. Sci. OA 2021, 7, 4. [Google Scholar] [CrossRef]
  30. Rigo, F.K.; Trevisan, G.; Godoy, M.C.; Rossato, M.F.; Dalmolin, G.D.; Silva, M.A.; Menezes, M.S.; Caumo, W.; Ferreira, J. Management of Neuropathic Chronic Pain with Methadone Combined with Ketamine: A Randomized, Double Blind, Active-Controlled Clinical Trial. Pain Physician 2017, 20, 207–215. [Google Scholar] [CrossRef]
  31. Trafton, J.A.; Ramani, A. Methadone: A new old drug with promises and pitfalls. Curr. Pain Headache Rep. 2009, 13, 24–30. [Google Scholar] [CrossRef]
  32. Eisenberg, E.; Pud, D. Can patients with chronic neuropathic pain be cured by acute administration of the NMDA receptor antagonist amantadine? Pain 1998, 74, 337–339. [Google Scholar] [CrossRef] [PubMed]
  33. Pud, D.; Eisenberg, E.; Spitzer, A.; Adler, R.; Fried, G.; Yarnitsky, D. The NMDA receptor antagonist amantadine reduces surgical neuropathic pain in cancer patients: A double blind, randomized, placebo controlled trial. Pain 1998, 75, 349–354. [Google Scholar] [CrossRef] [PubMed]
  34. Kleinbohl, D.; Gortelmeyer, R.; Bender, H.J. Amantadine sulfate reduces experimental sensitization and pain in chronic back pain patients. Anesth. Analg. 2006, 102, 840–847. [Google Scholar] [CrossRef] [PubMed]
  35. Bujak-Giżycka, B.; Kącka, K.; Suski, M.; Olszanecki, R.; Madej, J.; Dobrogowski, J.; Korbut, R. Beneficial effect of amantadine on Reduction of postoperative pain and morphine consumption in patients undergoing elective spine surgery. Pain Med. 2012, 13, 459–465. [Google Scholar] [CrossRef] [PubMed]
  36. Kubera, M.; Basta-Kaim, A.; Budziszewska, B.; Rogóż, Z.; Skuza, G.; Leśkiewicz, M.; Tetich, M.; Jaworska-Feil, L.; Maes, M.; Lasoń, W. Effect of amantadine and imipramine on immunological parameters of rats subjected to a forced swimming test. Int. J. Neuropsychopharmacol. 2006, 9, 297–305. [Google Scholar] [CrossRef] [PubMed]
  37. Tan, L.; Ge, H.; Tang, J.; Fu, C.; Duanmu, W.; Chen, Y.; Feng, H. Amantadine preserves dopamine level and attenuates depression-like behavior induced by traumatic brain injury in rats. Behav. Brain Res. 2015, 279, 274–282. [Google Scholar] [CrossRef] [PubMed]
  38. Müller, T. GOCOVRI® (amantadine) extended-release Capsules in Parkinson’s Disease. Neurodegener. Dis. Manag. 2022, 12, 15–28. [Google Scholar] [CrossRef]
  39. Lascelles, B.; Gaynor, J.; Smith, E.; Roe, S.; Marcellin-Little, D.; Davidson, G.; Boland, E.; Carr, J. Amantadine in a Multimodal Analgesic Regimen for Alleviation of Refractory Osteoarthritis Pain in Dogs. J. Vet. Intern. Med. 2008, 22, 53–59. [Google Scholar] [CrossRef] [PubMed]
  40. Coffeen, U.; López-Ávila, A.; Pellicer, F. Systemic amantadine decreases inflammatory and neuropathic nociception in the rat. Ment. Health 2009, 32, 139–144. [Google Scholar]
  41. Zhang, J.; Tan, H.; Jiang, W.; Zuo, Z. Amantadine Alleviates Postoperative Cognitive Dysfunction Possibly by Increasing Glial Cell Line-derived Neurotrophic Factor in Rats. Anesthesiology 2014, 121, 773–785. [Google Scholar] [CrossRef] [PubMed]
  42. Gottschalk, A.; Smith, D.S. New concepts in acute pain therapy: Preemptive analgesia. Am. Fam. Physician 2001, 15, 1979–1984. [Google Scholar] [PubMed]
  43. Fumagalli, G.; Monza, L.; Cavaletti, G.; Rigolio, R.; Meregalli, C. Neuroinflammatory Process Involved in Different Preclinical Models of Chemotherapy-Induced Peripheral Neuropathy. Front Immunol. 2021, 11, 626687. [Google Scholar] [CrossRef]
  44. Zhou, Y.Q.; Liu, Z.; Liu, Z.H.; Chen, S.P.; Li, M.; Shahveranov, A.; Tian, Y.K. Interleukin-6: An emerging regulator of pathological pain. J. Neuroinflamm. 2016, 13, 141. [Google Scholar] [CrossRef] [PubMed]
  45. Melemedjian, O.K.; Tillu, D.V.; Moy, J.K.; Asiedu, M.N.; Mandell, E.K.; Ghosh, S.; Price, T.J. Local Translation and Retrograde Axonal Transport of CREB Regulates IL-6-Induced Nociceptive Plasticity. Mol. Pain 2014, 10, 45. [Google Scholar] [CrossRef] [PubMed]
  46. Obreja, O.; Schmelz, M.; Poole, S.; Kress, M. Interleukin-6 in combination with its soluble IL-6 receptor sensitises rat skin nociceptors to heat, in vivo. Pain 2002, 96, 57–62. [Google Scholar] [CrossRef] [PubMed]
  47. Brenn, D.; Richter, F.; Schaible, H.G. Sensitization of unmyelinated sensory fibers of the joint nerve to mechanical stimuli by interleukin-6 in the rat: An inflammatory mechanism of joint pain. Arthritis Rheumatol. 2007, 56, 351–359. [Google Scholar] [CrossRef] [PubMed]
  48. Vazquez, E.; Kahlenbach, J.; Segond von Banchet, G.; König, C.; Schaible, H.G.; Ebersberger, A. Spinal interleukin-6 is an amplifier of arthritic pain in the rat. Arthritis Rheumatol. 2012, 64, 2233–2242. [Google Scholar] [CrossRef] [PubMed]
  49. Cunha, F.Q.; Poole, S.; Lorenzetti, B.B.; Ferreira, S.H. The pivotal role of tumor necrosis factor alpha in the development of inflammatory hyperalgesia. Br. J. Pharmacol. 1992, 107, 660–664. [Google Scholar] [CrossRef] [PubMed]
  50. Morgan, M.J.; Kim, Y.S.; Liu, Z.G. TNFalpha and reactive oxygen species in necrotic cell death. Cell Res. 2008, 18, 343–349. [Google Scholar] [CrossRef] [PubMed]
  51. Stellwagen, D.; Beattie, E.C.; Seo, J.Y.; Malenka, R.C. Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-alpha. J. Neurosci. 2005, 25, 3219–3228. [Google Scholar] [CrossRef] [PubMed]
  52. Dumont, A.O.; Goursaud, S.; Desmet, N.; Herman, S.E. Differential regulation of glutamate transporter subtypes by pro-inflammatory cytokine TNF-alpha in cortical astrocytes from a rat model of amyotrophic lateral sclerosis. PLoS ONE 2014, 9, e97649. [Google Scholar] [CrossRef] [PubMed]
  53. Clark, I.A.; Vissel, B. Excess cerebral TNF causing glutamate excitotoxicity rationalizes treatment of neurodegenerative diseases and neurogenic pain by anti-TNF agents. J. Neuroinflamm. 2016, 13, 236. [Google Scholar] [CrossRef] [PubMed]
  54. Park, C.; Lü, N.; Xu, Z.; Liu, T.; Serhan, C.; Ji, R. Resolving TRPV1- and TNF-α-mediated spinal cord synaptic plasticity and inflammatory pain with neuroprotectin D1. J. Neurosci. 2011, 31, 15072–15085. [Google Scholar] [CrossRef] [PubMed]
  55. Echeverry, S.; Wu, Y.; Zhang, J. Selectively reducing cytokine/chemokine expressing macrophages in injured nerves impairs the development of neuropathic pain. Neurol Exp. 2012, 240, 205–218. [Google Scholar] [CrossRef] [PubMed]
  56. Kiguchi, N.; Kobayashi, Y.; Maeda, T.; Saika, F.; Kishioka, S. CC-chemokine MIP-1α in the spinal cord contributes to nerve injury-induced neuropathic pain. Neurosci. Lett. 2010, 484, 17–21. [Google Scholar] [CrossRef] [PubMed]
  57. Dogan, G.; Karaca, O. N-methyl-D-aspartate receptor antagonists may ameliorate spinal cord injury by inhibiting oxidative stress: An experimental study in rats. Turk. Neurosurg. 2020, 30, 60–68. [Google Scholar] [CrossRef]
  58. Xing, W.; Huang, P.; Lu, Y.; Zeng, W.; Zuo, Z. Amantadine attenuates sepsis-induced cognitive dysfunction possibly not through inhibiting toll-like receptor 2. J. Mol. Med. 2018, 96, 391–402. [Google Scholar] [CrossRef]
  59. Milligan, E.D.; Sloane, E.M.; Langer, S.J.; Hughes, T.S.; Jekich, B.M.; Frank, M.G. Repeated intrathecal injections of plasmid DNA encoding interleukin-10 produce prolonged reversal of neuropathic pain. Pain 2006, 126, 294–308. [Google Scholar] [CrossRef] [PubMed]
  60. Chen, X.J.; Wang, L.; Song, X.Y. Mitoquinone alleviates vincristine-induced neuropathic pain through inhibiting oxidative stress and apoptosis via the improvement of mitochondrial dysfunction. Biomed. Pharmacother. 2020, 125, 110003. [Google Scholar] [CrossRef] [PubMed]
  61. Clark, A.K.; Malcangio, M. Fractalkine/CX3CR1 signaling during neuropathic pain. Front. Cell. Neurosci. 2014, 8, 121. [Google Scholar] [CrossRef] [PubMed]
  62. Döring, Y.; Pawig, L.; Weber, C.; Noels, H. The CXCL12/CXCR4 chemokine ligand/receptor axis in cardiovascular disease. Front. Physiol. 2014, 5, 212. [Google Scholar] [CrossRef] [PubMed]
  63. Bai, L.; Wang, X.; Li, Z.; Kong, C.; Zhao, Y.; Qian, J.L.; Kan, Q.; Zhang, W.; Xu, J.T. Upregulation of Chemokine CXCL12 in the Dorsal Root Ganglia and Spinal Cord Contributes to the Development and Maintenance of Neuropathic Pain Following Spared Nerve Injury in Rats. Neurosci. Bull. 2016, 32, 27–40. [Google Scholar] [CrossRef] [PubMed]
  64. Deng, L.; Guindon, J.; Vemuri, V.K.; Thakur, G.A.; White, F.A.; Makriyannis, A.; Hohmann, A.G. The maintenance of cisplatin-and paclitaxel-induced mechanical and cold allodynia is suppressed by cannabinoid CB2 receptor activation and independent of CXCR4 signaling in models of chemotherapy-induced peripheral neuropathy. Mol. Pain 2012, 8, 1744–8069. [Google Scholar] [CrossRef] [PubMed]
  65. Kesik, V.; Kurt, B.; Tunc, T.; Karslioglu, Y.; Citak, E.C.; Kismet, E.; Koseoglu, V. Adrenomedullin worsens skin necrosis in rats subjected to vincristine-induced extravasation. Clin. Exp. Dermatol. 2010, 35, 897–901. [Google Scholar] [CrossRef] [PubMed]
  66. Carozzi, V.A.; Canta, A.; Chiorazzi, A. Chemotherapy-induced peripheral neuropathy: What do we know about mechanisms? Neurosci. Lett. 2015, 596, 90–107. [Google Scholar] [CrossRef] [PubMed]
  67. Kelley, M.R.; Fehrenbacher, J.C. Challenges and opportunities identifying therapeutic targets for chemotherapy-induced peripheral neuropathy resulting from oxidative DNA damage. Neural Regen. Res. 2017, 12, 72–74. [Google Scholar] [CrossRef] [PubMed]
  68. Shim, H.S.; Bae, C.; Wang, J.; Lee, K.H.; Hankerd, K.M.; Kim, H.K.; La, J.H. Peripheral and central oxidative stress in chemotherapy-induced neuropathic pain. Mol. Pain 2019, 15, 1744806919840098. [Google Scholar] [CrossRef] [PubMed]
  69. Areti, A.; Yerra, V.G.; Naidu, V.; Kumar, A. Oxidative stress and nerve damage: Role in chemotherapy induced peripheral neuropathy. Redox Biol. 2014, 2, 289–295. [Google Scholar] [CrossRef] [PubMed]
  70. Culotta, V.S. Superoxide dismutase, oxidative stress, and cell metabolism. Curr. Top. Cell. Regul. 2001, 36, 117–132. [Google Scholar] [CrossRef] [PubMed]
  71. Xie, Y.; Mu, H.; Li, Z.; Ma, J.; Wang, Y. Suppression of chronic central pain by superoxide dismutase in rats with spinal cord injury: Inhibition of the NMDA receptor implicated. Exp. Ther. Med. 2014, 8, 1137–1141. [Google Scholar] [CrossRef] [PubMed]
  72. Orhan, M.; Taş Tuna, A.; Ünal, Y.; Arslan, M.; Yazar, H.; Sezen, Ş.C.; Gözükara, S.I.; Palabiyik, O. The effects of amantadine on lung tissue in lower limb ischemia/reperfusion injury model in rats. Turk. J. Thorac. Cardiovasc. Surg. 2021, 29, 77–83. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, Y.; Wang, S.; Wang, Z.; Ding, M.; Li, X.; Guo, J.; Han, G.; Zhao, P. Dexmedetomidine Alleviated Endoplasmic Reticulum Stress via Inducing ER-phagy in the Spinal Cord of Neuropathic Pain Model. Front. Neurosci. 2020, 14, 90. [Google Scholar] [CrossRef] [PubMed]
  74. Seo, E.-H.; Piao, L.; Cho, E.-H.; Hong, S.-W.; Kim, S.-H. The Effect of Ketamine on Endoplasmic Reticulum Stress in Rats with Neuropathic Pain. Int. J. Mol. Sci. 2023, 24, 5336. [Google Scholar] [CrossRef] [PubMed]
  75. Batinic-Haberle, I.; Tovmasyan, A.; Spasojevic, I. Mn 1 Porphyrin-Based Redox-Active Drugs: Differential Effects as Cancer Therapeutics and Protectors of Normal Tissue against Oxidative Injury. Antioxid. Redox Signal. 2018, 29, 1691–1724. [Google Scholar] [CrossRef] [PubMed]
  76. Portt, L.; Norman, G.; Clapp, C.; Greenwood, M.; Greenwood, M.T. Anti-apoptosis and cell survival: A review. Biochem. Biophys. Acta 2010, 1813, 238–259. [Google Scholar] [CrossRef]
  77. Ahmed, S.; Ahmed, E.M.; Hantash, S.; Zakaria, S. Potential of Amantadine to Ameliorate Glutamate-Induced Pyramidal Cells Toxicity in Juvenile Rat’ Brain Cortex. Neurotox. Res. 2021, 39, 1203–1210. [Google Scholar] [CrossRef]
Figure 1. Assessment of the nociceptive mechanical threshold (g) using a digital analgesimeter, performed daily for 14 days. Values represent mean ± SEM. # (differs GPC, A2, A5, A12, A25 e A50; p < 0.05). * (differs GNC, naïve, A2, A5 and GPC; p < 0.05).
Figure 1. Assessment of the nociceptive mechanical threshold (g) using a digital analgesimeter, performed daily for 14 days. Values represent mean ± SEM. # (differs GPC, A2, A5, A12, A25 e A50; p < 0.05). * (differs GNC, naïve, A2, A5 and GPC; p < 0.05).
Animals 14 01941 g001
Figure 2. Expression of IL-6, IL-10, TNF-α and MIP-1α in the spinal cord of rats treated with amantadine for vincristine-induced peripheral neuropathy. (AE) Photomicrographs of immunohistochemical expression of IL6 (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (GK) Photomicrographs of the immunohistochemical expression of IL10 (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (MQ) Photomicrographs of the immunohistochemical expression of TNF-α (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (SX) Photomicrographs of the immunohistochemical expression of MIP1a (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (F) Immunolabeling area, in pixels, of IL6 expression (mean ± SEM; n = 6). (L) Immunolabeling area, in pixels, of IL-10 expression (mean ± SEM; n = 6). (R) Immunolabeling area, in pixels, of TNF-α expression (mean ± SEM; n = 6). (Z) Immunolabeling area, in pixels, of MIP-1α expression (mean ± SEM; n = 6). (WY2) Relative gene expression of IL-6, IL-10, IL-18 and IL-1β in the spinal cord (mean ± SEM; n = 6). * (p < 0.05), ** (p < 0.01), *** (p < 0.001), **** (p < 0.0001).
Figure 2. Expression of IL-6, IL-10, TNF-α and MIP-1α in the spinal cord of rats treated with amantadine for vincristine-induced peripheral neuropathy. (AE) Photomicrographs of immunohistochemical expression of IL6 (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (GK) Photomicrographs of the immunohistochemical expression of IL10 (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (MQ) Photomicrographs of the immunohistochemical expression of TNF-α (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (SX) Photomicrographs of the immunohistochemical expression of MIP1a (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (F) Immunolabeling area, in pixels, of IL6 expression (mean ± SEM; n = 6). (L) Immunolabeling area, in pixels, of IL-10 expression (mean ± SEM; n = 6). (R) Immunolabeling area, in pixels, of TNF-α expression (mean ± SEM; n = 6). (Z) Immunolabeling area, in pixels, of MIP-1α expression (mean ± SEM; n = 6). (WY2) Relative gene expression of IL-6, IL-10, IL-18 and IL-1β in the spinal cord (mean ± SEM; n = 6). * (p < 0.05), ** (p < 0.01), *** (p < 0.001), **** (p < 0.0001).
Animals 14 01941 g002
Figure 3. Expression of CX3CR1 and CXCR4 in the spinal cord of rats treated with amantadine for vincristine-induced peripheral neuropathy. (AE) Photomicrographs of immunostaining expression in rat spinal cord of CX3CR1 (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (FJ) Photomicrographs of the immunohistochemical expression of CXCR4 (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (K) Immunolabeling area, in pixels, of CX3CR1 expression (mean ± SEM; n = 6). (L) Immunolabeling area, in pixels, of CXCR4 expression (mean ± SEM; n = 6). *** (p < 0.001), **** (p < 0.0001).
Figure 3. Expression of CX3CR1 and CXCR4 in the spinal cord of rats treated with amantadine for vincristine-induced peripheral neuropathy. (AE) Photomicrographs of immunostaining expression in rat spinal cord of CX3CR1 (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (FJ) Photomicrographs of the immunohistochemical expression of CXCR4 (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (K) Immunolabeling area, in pixels, of CX3CR1 expression (mean ± SEM; n = 6). (L) Immunolabeling area, in pixels, of CXCR4 expression (mean ± SEM; n = 6). *** (p < 0.001), **** (p < 0.0001).
Animals 14 01941 g003
Figure 4. Expression of CAT, SOD and GPx in the spinal cord of rats treated with amantadine for vincristine-induced peripheral neuropathy. (AE) Photomicrographs of immunostaining expression in rat spinal cord of catalase (CAT) (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (GK) Photomicrographs of immunostaining expression in rat spinal cord of Superoxide Dismutase (SOD) (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (MQ) Photomicrographs of immunostaining expression in rat spinal cord of Glutathione Peroxidase (GPx) (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (F) Immunolabeling area, in pixels, of CAT expression (mean ± SEM; n = 6). (L) Immunolabeling area, in pixels, of SOD expression (mean ± SEM; n = 6). (R) Immunolabeling area, in pixels, of GPx expression (mean ± SEM; n = 6). (S) Enzymatic activity expression of CAT (mean ± SEM; n = 6). (T) Enzymatic activity expression of SOD (mean ± SEM; n = 6). *** (p < 0.001), **** (p < 0.0001).
Figure 4. Expression of CAT, SOD and GPx in the spinal cord of rats treated with amantadine for vincristine-induced peripheral neuropathy. (AE) Photomicrographs of immunostaining expression in rat spinal cord of catalase (CAT) (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (GK) Photomicrographs of immunostaining expression in rat spinal cord of Superoxide Dismutase (SOD) (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (MQ) Photomicrographs of immunostaining expression in rat spinal cord of Glutathione Peroxidase (GPx) (streptavidin–biotin–peroxidase; Harris hematoxylin; 40×). (F) Immunolabeling area, in pixels, of CAT expression (mean ± SEM; n = 6). (L) Immunolabeling area, in pixels, of SOD expression (mean ± SEM; n = 6). (R) Immunolabeling area, in pixels, of GPx expression (mean ± SEM; n = 6). (S) Enzymatic activity expression of CAT (mean ± SEM; n = 6). (T) Enzymatic activity expression of SOD (mean ± SEM; n = 6). *** (p < 0.001), **** (p < 0.0001).
Animals 14 01941 g004
Figure 5. Relative gene expression graphs of Grp78, CHOP, Ho1 and Perk in quantitative PCR (qPCR) technique in rat spinal cord in control groups and groups treated with amantadine at doses of 25 mg and 50 mg. (A) Relative gene expression of Grp78 (mean ± SEM; n = 6); (B) relative gene expression of CHOP (mean ± SEM; n = 6); (C) relative gene expression of Ho1 (mean ± SEM; n = 6); and (D) relative gene expression of Perk (mean ± SEM; n = 6). * (p < 0.05).
Figure 5. Relative gene expression graphs of Grp78, CHOP, Ho1 and Perk in quantitative PCR (qPCR) technique in rat spinal cord in control groups and groups treated with amantadine at doses of 25 mg and 50 mg. (A) Relative gene expression of Grp78 (mean ± SEM; n = 6); (B) relative gene expression of CHOP (mean ± SEM; n = 6); (C) relative gene expression of Ho1 (mean ± SEM; n = 6); and (D) relative gene expression of Perk (mean ± SEM; n = 6). * (p < 0.05).
Animals 14 01941 g005
Figure 6. Relative gene expression graphs of Bax, Bcl xL, Casp 3 and Casp 9 in quantitative PCR (qPCR) technique in rat spinal cord in control groups and groups treated with amantadine at doses of 25 mg and 50 mg. (A) Relative gene expression of Bax (mean ± SEM; n = 6); (B) relative gene expression of Bcl xL (mean ± SEM; n = 6); (C) relative gene expression of Casp 3 (mean ± SEM; n = 6); and (D) relative gene expression of Casp 9 (mean ± SEM; n = 6). * (p < 0.05), ** (p < 0.01), *** (p < 0.001).
Figure 6. Relative gene expression graphs of Bax, Bcl xL, Casp 3 and Casp 9 in quantitative PCR (qPCR) technique in rat spinal cord in control groups and groups treated with amantadine at doses of 25 mg and 50 mg. (A) Relative gene expression of Bax (mean ± SEM; n = 6); (B) relative gene expression of Bcl xL (mean ± SEM; n = 6); (C) relative gene expression of Casp 3 (mean ± SEM; n = 6); and (D) relative gene expression of Casp 9 (mean ± SEM; n = 6). * (p < 0.05), ** (p < 0.01), *** (p < 0.001).
Animals 14 01941 g006
Table 1. Antibodies used in immunohistochemistry and respective dilutions, development time and code.
Table 1. Antibodies used in immunohistochemistry and respective dilutions, development time and code.
AntibodyDilutionDAB TimeCode
Anti-CX3CR11:25010 minanti-CX3CR1, sc-377227
Anti-CXCR-4/IgC1:2005 minanti-CXCR-4, sc-53534
Anti-Catalase1:2007 minanti-CAT, sc-271803
Anti-GPx1:20010 minanti-GPx, sc-133152
Anti-IL-101:25003 minanti-IL10, sc-365858
Anti-IL-61:15003 minanti-IL6, sc-28343
Anti-MIP-1α1:10012 minanti-MIP-1α, sc-36569
Anti-SOD1:800015 santi-SOD, sc-101523
Anti-TNF-α1:5003 minanti-TNFα, sc-33639
Table 2. List of genes and nucleotide sequences of primers for qRT-PCR.
Table 2. List of genes and nucleotide sequences of primers for qRT-PCR.
GenesStartersNoAccess
Grp78Forward: TGAAGGGGAGCGTCTGATTG
Reverse: TCATTCCAAGTGCGTCCGAT
NM_013083.2
ChopForward: TGGCACAGCTTGCTGAAGAG
Reverse: TCAGGCGCTCGATTTCCT
NM_001109986.1
PerkForward: GGCTGGTGAGGGATGGTAAA
Reverse: TTGGCTGTGTAACTTGTGTCATC
NM_031599.2
Ho1Forward: CAGCATACGTAAAGCGTCTCCA
Reverse: CATGGCCTTCTGCGCAATCTTCTT
NM_012580.2
BaxForward: GCACGTCTGCGGGAG
Reverse: ATCTGTTCAGAGCTGGTGGG
NM_017059.2
Bcl-xlForward: AGAACCTGGACTCAGACCTTC
Reverse: TCCAGGATCCAAAGCCAAGA
XM_039104291.1
CASP 3Forward: GAGCTTGGAACGCGAAGAAA
Reverse: AGTCCATCGACTTGCTTCCA
NM_012922.2
CASP 9Forward: TCCCCACTGATCAAGTCTCCT
Reverse: CCAGGCTCACTTAGCAAGGAA
NM_031632.2
IL-6Forward: GACTTCCAGCCAGTTGCCTTR
Reverse: AAGTCTCCTCTCCGGACTTGT
NM_053595.2
IL-1βForward: GCACAGTTCCCCCAACTGGTA
Reverse: TGTCCCGACCATTGCTGTTT
NM_ 031512.2
IL-10Forward: ACCACTTTGGCAGACTTCCT
Reverse: ACACAGGCGGGTTTCTTTTG
NM_053595.2
IL-18Forward: GACTTCCAGCCAGTTGCCTTR
Reverse: AAGTCTCCTCTCCGGACTTGT
NM_019174.4
GapdhForward: GCGCTACAGCGGATTTTTGA
Reverse: GAAGGCATACACGGTGGACT
NM_031797.2
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Drummond, I.S.A.; de Oliveira, J.N.S.; Niella, R.V.; Silva, Á.J.C.; de Oliveira, I.S.; de Souza, S.S.; da Costa Marques, C.S.; Corrêa, J.M.X.; Silva, J.F.; de Lavor, M.S.L. Evaluation of the Therapeutic Potential of Amantadine in a Vincristine-Induced Peripheral Neuropathy Model in Rats. Animals 2024, 14, 1941. https://doi.org/10.3390/ani14131941

AMA Style

Drummond ISA, de Oliveira JNS, Niella RV, Silva ÁJC, de Oliveira IS, de Souza SS, da Costa Marques CS, Corrêa JMX, Silva JF, de Lavor MSL. Evaluation of the Therapeutic Potential of Amantadine in a Vincristine-Induced Peripheral Neuropathy Model in Rats. Animals. 2024; 14(13):1941. https://doi.org/10.3390/ani14131941

Chicago/Turabian Style

Drummond, Isabela Santana Albertazzi, Jéssica Natália Silva de Oliveira, Raquel Vieira Niella, Álvaro José Chávez Silva, Iago Santos de Oliveira, Sophia Saraiva de Souza, Claire Souza da Costa Marques, Janaina Maria Xavier Corrêa, Juneo Freitas Silva, and Mário Sérgio Lima de Lavor. 2024. "Evaluation of the Therapeutic Potential of Amantadine in a Vincristine-Induced Peripheral Neuropathy Model in Rats" Animals 14, no. 13: 1941. https://doi.org/10.3390/ani14131941

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop