Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Int J Pharm ; 640: 123037, 2023 Jun 10.
Article En | MEDLINE | ID: mdl-37172632

Nanotechnology plays an integral role in multimodal analgesia. In this study, we co-encapsulated metformin (Met) and curcumin (Cur) into chitosan/alginate (CTS/ALG) nanoparticles (NPs) at their synergistic drug ratio by applying response surface methodology. The optimized Met-Cur-CTS/ALG-NPs were achieved with Pluronic® F-127 2.33 % (w/v), Met 5.91 mg, and CTS:ALG mass ratio 0.05:1. The prepared Met-Cur-CTS/ALG-NPs had 243 nm particle size, -21.6 mV zeta potential, 32.6 and 44.2 % Met and Cur encapsulations, 19.6 and 6.8 % Met and Cur loading, respectively, and 2.9:1 Met:Cur mass ratio. Met-Cur-CTS/ALG-NPs displayed stability under simulated gastrointestinal (GI) fluid conditions and during storage. In vitro release study of Met-Cur-CTS/ALG-NPs in simulated GI fluids showed sustained release, with Met exhibiting Fickian diffusion and Cur demonstrating non-Fickian diffusion following the Korsmeyer-Peppas model. Met-Cur-CTS/ALG-NPs exhibited increased mucoadhesion and improved cellular uptake in Caco-2 cells. Additionally, Met-Cur-CTS/ALG-NPs exhibited better anti-inflammatory effects in lipopolysaccharide-stimulated RAW 264.7 macrophage and BV-2 microglial cells than the equivalent amount of the Met-Cur physical mixture, indicating a greater ability to modulate peripheral and central immune mechanisms of pain. In the mouse formalin-induced pain model, Met-Cur-CTS/ALG-NPs administered orally exhibited better attenuation of pain-like behaviors and proinflammatory cytokine release compared to the Met-Cur physical mixture. Furthermore, Met-Cur-CTS/ALG-NPs did not induce significant side effects in mice at therapeutic doses. Altogether, the present study establishes a CTS/ALG nano-delivery system for Met-Cur combination against pain with improved efficacy and safety.


Chitosan , Curcumin , Metformin , Nanoparticles , Humans , Mice , Animals , Drug Carriers , Curcumin/pharmacology , Chitosan/pharmacology , Caco-2 Cells , Alginates/pharmacology , Particle Size
2.
Int J Mol Sci ; 24(9)2023 Apr 29.
Article En | MEDLINE | ID: mdl-37175777

Bacterial meningitis remains one of the most prevalent infectious diseases worldwide. Although advances in medical care have improved mortality and morbidity, neurological complications remain high. Therefore, aside from antibiotics, therapeutic adjuvants targeting neuroinflammation are essential to combat the long-term neuronal sequelae of bacterial meningitis. In the present study, we propose (-)-dendroparishiol as a potential add-on therapy to improve neuroinflammation associated with bacterial meningitis. The biological activity of (-)-dendroparishiol was first predicted by computational analysis and further confirmed in vitro using a cell-based assay with LPS-induced BV-2 microglial cells. Biological pathways involved with (-)-dendroparishiol were identified by applying network pharmacology. Computational predictions of biological activity indicated possible attenuation of several inflammatory processes by (-)-dendroparishiol. In LPS-induced BV-2 microglial cells, (-)-dendroparishiol significantly reduced the expression of inflammatory mediators: iNOS, NO, COX-2, IL-6, and TNF-α. Molecular docking results demonstrated the potential iNOS and COX-2 inhibitory activity of (-)-dendroparishiol. Network pharmacological analysis indicated the plausible role of (-)-dendroparishiol in biological processes involved in oxidative stress and neuroinflammation with enrichment in neuroinflammatory pathways. Overall, this study provides scientific evidence for the potential application of (-)-dendroparishiol in the management of bacterial meningitis-associated neuroinflammation.


Inflammation , Meningitis, Bacterial , Humans , Inflammation/metabolism , Neuroinflammatory Diseases , Lipopolysaccharides/adverse effects , Molecular Docking Simulation , Network Pharmacology , Microglia/metabolism , Meningitis, Bacterial/metabolism , NF-kappa B/metabolism
3.
RSC Adv ; 13(16): 10757-10767, 2023 Apr 03.
Article En | MEDLINE | ID: mdl-37025673

Central nervous system (CNS) diseases are a significant health burden globally, with the development of novel drugs lagging behind clinical needs. Orchidaceae plants have been traditionally used to treat CNS diseases, leading to the identification of therapeutic leads against CNS diseases from the Aerides falcata orchid plant in the present study. The study isolated and characterized ten compounds, including a previously undescribed biphenanthrene derivative, Aerifalcatin (1), for the first time from the A. falcata extract. The novel compound 1 and known compounds, such as 2,7-dihydroxy-3,4,6-trimethoxyphenanthrene (5), agrostonin (7), and syringaresinol (9), showed potential activity in CNS-associated disease models. Notably, compounds 1, 5, 7, and 9 demonstrated the ability to alleviate LPS-induced NO release in BV-2 microglial cells, with IC50 values of 0.9, 2.5, 2.6, and 1.4 µM, respectively. These compounds also significantly inhibited the release of pro-inflammatory cytokines, IL-6 and TNF-α, reflecting their potential anti-neuroinflammatory effects. Additionally, compounds 1, 7, and 9 were found to reduce cell growth and migration of glioblastoma and neuroblastoma cells, indicating their potential use as anticancer agents in the CNS. In summary, the bioactive agents isolated from the A. falcata extract offer plausible therapeutic options for CNS diseases.

4.
J Tradit Complement Med ; 13(1): 81-92, 2023 Jan.
Article En | MEDLINE | ID: mdl-36685077

Background: Clerodendrum petasites, an herbal plant in Thailand, has been used for many years in folk medicine. However, scientific evidence regarding CNS safety pharmacology and antinociceptive activity of C. petasites (CP) has not yet been well characterized. Purpose: The present study aimed to assess the CNS safety pharmacology and antinociceptive and antiinflammatory effects of CP extract. Methods: The effect of CP extract on CNS safety pharmacology was assessed using LABORAS automated home cage monitoring and rotarod test. Its pharmacological activity was evaluated both in-vitro, and in-vivo using hot-plate, acetic acid-induced writhing, formalin, and carrageenan-induced paw edema models. Results and conclusion: CP extract significantly improved thermal and chemical nociceptive behaviors and acute inflammatory pain at all doses: 300, 600, and 1200 mg/kg, p.o. The antiinflammatory effect of CP extract in inflammatory pain models was comparable to the effect of positive control: indomethacin 10 mg/kg at all dose levels tested. Further, the CP extract at 600 mg/kg dose significantly inhibited 82.3% of carrageenan-induced total edema. In-vitro, CP extract at 12.5, 25, and 50 µg/mL concentrations significantly reduced the expression of LPS-induced nitric oxide, IL-6, and TNF-α expression in both RAW 264.7 macrophage and BV-2 microglial cell lines. In addition, CP extract did not show any potential effects on the CNS, indicated by no significant effects on motor coordination, spontaneous locomotor activity, general behaviors, and well-being compared to vehicle-treated mice (p > 0.05). Overall, the present study evidences the potential antinociceptive, antiinflammatory efficacies of CP extract with a favorable CNS safety profile.

5.
In Vivo ; 36(6): 2730-2739, 2022.
Article En | MEDLINE | ID: mdl-36309371

BACKGROUND/AIM: Fine particulate matter (PM2.5) in air pollution causes skin damage through the induction of oxidative stress in the epidermis. Antioxidants help counteract cellular oxidant species and maintain cell homeostasis. This study aimed to examine the protective effect of standardized ethanolic extract of Thunbergia laurifolia leaves on PM2.5-mediated oxidative stress in epidermal keratinocytes. MATERIALS AND METHODS: The extract was standardized with rosmarinic acid. Effects of standardized T. laurifolia extract (STLE) (0-400 µg/ml) and PM2.5 (0-32 µg/ml) on cell viability after 24 h of treatment were detected using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. PM2.5 (0-32 µg/ml) induction of intracellular reactive oxygen species (ROS) at 6 h was monitored using 2',7'-dichlorodihydrofluorescein diacetate. Cells were co-treated for 6 h with PM2.5 (32 µg/ml) and STLE (25-100 µg/ml) and monitored for oxidative stress inhibition. Proteins related to cellular antioxidant defense system were examined by western blot analysis, after co-treatment and STLE treatment for 6 h and 24 h, respectively. Nuclear expression of nuclear factor erythroid 2-related factor (NRF2) and p62 were determined by immunofluorescence after co-treatment of 6 h. RESULTS: PM2.5 (32 µg/ml) remarkably induced ROS production within 6 h. The co-treatment dramatically inhibited PM2.5-induced oxidative stress at 6 h. In addition, STLE enhanced cellular defense system by increasing the levels of p62, NRF2 and superoxide dismutase 1 proteins. STLE stimulated nuclear localization and function of NRF2 and p62 proteins, while suppressing Kelch-like ECH-associated protein 1. CONCLUSION: STLE exhibits promising natural antioxidant activity against oxidative stress induced by PM2.5 in keratinocytes.


Acanthaceae , NF-E2-Related Factor 2 , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Oxidative Stress , Signal Transduction , Antioxidants/pharmacology , Antioxidants/metabolism , Particulate Matter/adverse effects , Acanthaceae/metabolism
6.
Chin Med ; 17(1): 119, 2022 Oct 23.
Article En | MEDLINE | ID: mdl-36274168

BACKGROUND: Curcumin and piperine are major bioactive compounds of Curcuma longa and Piper nigrum, widely consumed as spices and flock medicine. The combinational use of these plants is a common practice in Southeast Asia. Synergism between curcumin and piperine has been found in several animal models but not in periodontal disease and diabetes, and the antinociceptive interaction is still unknown. Hence, the present study aimed to assess the interaction between curcumin and piperine in pain and its potential CNS side effect profile. METHODS: Formalin test and in vitro LPS-stimulated RAW 264.7 macrophage cells were used to assess the synergistic interaction of curcumin and piperine in a mouse model of inflammatory pain. Tail-flick and cold plate tests were applied to determine the antinociceptive synergism between piperine and curcumin. The interaction was determined by applying isobolographic analysis. The potential CNS-side effects of the curcumin and piperine combination were also assessed using LABORAS automated home-cage behavioral analysis. RESULTS: Curcumin alone dose-dependently improved pain-like behaviors in the formalin, tail-flick, and cold plate tests with the ED50 of 71.4, 34.4, and 31.9 mg/kg, respectively. Additionally, piperine exhibited efficacy in the formalin, tail-flick, and cold plate tests with the ED50 of 18.4, 8.1, and 28.1 mg/kg, respectively. The combination of curcumin and piperine (1:1 ED50 ratio) produced synergistic interaction in the formalin, tail-flick, and cold plate tests as assessed significantly lower experimental ED50 values (5.9, 5.2, and 5.5 mg/kg) compared to theoretical ED50 values (44.9, 21.3, and 30.0 mg/kg), isobologram analysis, and interaction index values of 0.13, 0.24 and 0.18, respectively. The synergistic interaction of curcumin and piperine was further confirmed by the efficacy of the combination in LPS-stimulated RAW 264.7 macrophage cells. Curcumin and piperine interacted synergistically, reducing proinflammatory mediators. The combination also demonstrated better compatibility profiles with neuronal cells. Furthermore, the curcumin-piperine combination had no effects on mouse spontaneous locomotor behaviors in LABORAS automated home cage monitoring. CONCLUSION: Overall, the present study demonstrates strong antinociceptive synergism between curcumin and piperine in mouse models with no potential CNS side effects, suggesting its possible use in clinical trials.

7.
ACS Pharmacol Transl Sci ; 5(9): 774-790, 2022 Sep 09.
Article En | MEDLINE | ID: mdl-36110378

Curcumin is a naturally occurring polyphenol compound with potential analgesic effects. It has been shown to improve pain-like behaviors in numerous models of pain. Despite its potential, curcumin exhibits poor physicochemical and pharmacokinetic properties, which hinder its oral therapeutic efficacy. Curcumin diethyl γ-aminobutyrate (CUR-2GE), a carbamate prodrug of curcumin, was designed to overcome these limitations and demonstrated greater anti-neuroinflammatory effects compared to curcumin in vitro. Thus, this study evaluated the effect of CUR-2GE and its parent compound on pain-like behaviors in carrageenan- and LPS-induced mouse models. The possible side effects of CUR-2GE were also assessed by exploring its effects on motor coordination and spontaneous locomotor activity after acute and chronic treatments. The results showed that CUR-2GE improved mechanical and thermal hyperalgesia and locomotor activity to a greater extent than curcumin in carrageenan-induced mice. These results are in line with the ability of CUR-2GE to suppress peripheral inflammation in the paw tissue of carrageenan-induced mice, indicated by a significant decrease in TNF-α and IL-6 expression levels. Similarly, in LPS-induced mice, CUR-2GE improved sickness and pain-like behaviors (exploratory behaviors and long-term locomotor activity) to a greater extent than curcumin. Furthermore, CUR-2GE significantly reduced the level of proinflammatory cytokines in both the plasma and spinal cord tissue of LPS-induced mice, exhibiting significantly higher inhibition than curcumin. Moreover, the motor coordination, and locomotive behaviors of mice were not affected by both acute and chronic administration of CUR-2GE, indicating no potential CNS side effects. Thus, CUR-2GE demonstrated enhanced therapeutic efficacy in mouse models of inflammatory pain without any possible CNS side effects, suggesting its potential to be developed as an analgesic agent against inflammatory pain.

8.
J Nat Prod ; 85(7): 1816-1825, 2022 07 22.
Article En | MEDLINE | ID: mdl-35707966

Batatasin III is a stilbenoid compound present in a wide variety of Dendrobium species. Although the pharmacological efficacy of batatasin III has been reported in several disease models, its antinociceptive efficacy and central nervous system (CNS) side effects remain unknown. Thus, this study examined the effects of batatasin III on pain-like behaviors in mouse models of formalin- and lipopolysaccharide (LPS)-induced inflammatory pain. The results revealed a significant antinociceptive effect of batatasin III in both models, as 50 mg/kg batatasin III elicited comparable antinociception as 10 mg/kg indomethacin. Further, the anti-inflammatory effect of batatasin III was assessed in LPS-induced RAW 264.7 macrophages and BV-2 microglial cells. The compound significantly reduced the levels of inflammatory mediators (nitric oxide, TNF-α, and IL-6) in LPS-stimulated cells in a concentration-dependent manner. Following efficacy evaluations, the potential CNS side effects of batatasin III were evaluated using the rotarod test and the Laboratory Animal Behavior Observation, Registration, and Analysis System. Batatasin III-treated mice exhibited comparable forced, spontaneous, and general locomotive behaviors to vehicle-treated mice, indicating no potential CNS side effects. Overall, this study demonstrated the preclinical antinociceptive efficacy and CNS safety of batatasin III, suggesting its potential role in the development of new analgesics.


Dendrobium , Stilbenes , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Lipopolysaccharides/pharmacology , Mice , Nitric Oxide , Pain/chemically induced , Pain/drug therapy
9.
Sci Rep ; 12(1): 9713, 2022 06 11.
Article En | MEDLINE | ID: mdl-35690654

Metformin is a well-tolerated antidiabetic drug and has recently been repurposed for numerous diseases, including pain. However, a higher dose of metformin is required for effective analgesia, which can potentiate its dose-dependent gastrointestinal side effects. Curcumin is a natural polyphenol and has beneficial therapeutic effects on pain. Curcumin has been used as an analgesic adjuvant with several analgesic drugs, allowing synergistic antinociceptive effects. Nevertheless, whether curcumin can exert synergistic analgesia with metformin is still unknown. In the present study, the nature of curcumin-metformin anti-inflammatory interaction was evaluated in in vitro using lipopolysaccharide-induced RAW 264.7 macrophage and BV-2 microglia cells. In both macrophage and microglia, curcumin effectively potentiates the anti-inflammatory effects of metformin, indicating potential synergistic effects in both peripheral and central pathways of pain. The nature of the interaction between curcumin and metformin was further recapitulated using a mouse model of formalin-induced pain. Coadministration of curcumin and metformin at a 1:1 fixed ratio of their ED50 doses significantly reduced the dose required to produce a 50% effect compared to the theoretically required dose in phase II of the formalin test with a combination index value of 0.24. Besides, the synergistic interaction does not appear to involve severe CNS side effects indicated by no motor alterations, no alterations in short-term and long-term locomotive behaviors, and the general well-being of mice. Our findings suggest that curcumin exerts synergistic anti-inflammation with metformin with no potential CNS adverse effects.


Curcumin , Metformin , Analgesics/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Curcumin/pharmacology , Curcumin/therapeutic use , Humans , Metformin/therapeutic use , Pain/drug therapy
10.
BMC Complement Med Ther ; 22(1): 139, 2022 May 18.
Article En | MEDLINE | ID: mdl-35585532

BACKGROUND: Crinum latifolium L. (Amaryllidaceae) has been used in Southeast Asian traditional medicine to alleviate the symptoms of benign prostatic hyperplasia (BPH). The pathological mechanism of BPH is associated with the induction of prostate stromal cell proliferation through transforming growth factor-beta (TGF-ß). Standardization as well as investigation of the potential anti-BPH activity of C. latifolium extract could benefit the further development of BPH-related analyses and provide evidence to support the application of this extract for BPH treatment. This study aimed to standardize and investigate the antiproliferative activity of the ethanolic extract of C. latifolium leaves. The major alkaloids isolated from C. latifolium were also explored for their potential use as bioactive markers. METHODS: Two major alkaloids were isolated from the ethanolic extract of C. latifolium leaves by chromatographic techniques, identified by NMR and MS, and quantified by a validated UHPLC method. Their antiproliferative activity was studied in human prostate stromal cells (WPMY-1) induced by TGF-ß. The synergistic effect of combining the two major isolated alkaloids was analyzed by the zero interaction potency (ZIP) model. RESULTS: Two alkaloids, lycorine (1) and 6α-hydroxybuphanidrine (2), were isolated from the ethanolic leaf extract of C. latifolium. A UHPLC method for the quantification of (1) and (2) was developed and validated in terms of linearity, precision, and accuracy. The C. latifolium leaf extract contained 0.279 ± 0.003% (1) and 0.232 ± 0.004% (2). The crude extract was more potent than either (1) and (2) alone against TGF-ß-treated WPMY-1 cell proliferation. The drug combination study revealed that the greatest synergistic effect of (1) and (2) was achieved at a 1:1 ratio. CONCLUSIONS: The results of this study support the anti-BPH activity of C. latifolium in traditional medicine and suggest that these the two isolated alkaloids may promote the efficacy of the C. latifolium extract. Additionally, major alkaloids (1) and (2) can be used as bioactive markers for the standardization of C. latifolium extracts.


Alkaloids , Crinum , Prostatic Hyperplasia , Alkaloids/pharmacology , Crinum/chemistry , Humans , Male , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Prostate/pathology , Prostatic Hyperplasia/drug therapy , Reference Standards , Stromal Cells/pathology , Transforming Growth Factor beta
11.
PLoS One ; 17(3): e0265689, 2022.
Article En | MEDLINE | ID: mdl-35303012

Curcumin is a polyphenol compound that alleviates several neuroinflammation-related diseases including Alzheimer's disease, Parkinson's disease, multiple sclerosis, epilepsy and cerebral injury. However, the therapeutic efficacy of curcumin is limited by its poor physicochemical properties. The present study aimed to develop a new carrier-linked curcumin prodrug, curcumin diethyl γ-aminobutyrate (CUR-2GE), with improved physicochemical and anti-neuroinflammatory properties. CUR-2GE was designed and synthesized by conjugating curcumin with gamma-aminobutyric acid ethyl ester (GE) via a carbamate linkage. The carbamate linkage was selected to increase stability at acidic pH while GE served as a promoiety for lipophilic enhancement. The synthesized CUR-2GE was investigated for solubility, partition coefficient, stability, and bioconversion. The solubility of CUR-2GE was less than 0.05 µg/mL similar to that of curcumin, while the lipophilicity with log P of 3.57 was significantly increased. CUR-2GE was resistant to chemical hydrolysis at acidic pH (pH 1.2 and 4.5) as anticipated but rapidly hydrolyzed at pH 6.8 and 7.4. The incomplete hydrolysis of CUR-2GE was observed in simulated gastrointestinal fluids which liberated the intermediate curcumin monoethyl γ-aminobutyric acid (CUR-1GE) and the parent curcumin. In plasma, CUR-2GE was sequentially converted to CUR-1GE and curcumin within 1 h. In lipopolysaccharide (LPS)-stimulated BV-2 microglial cells, CUR-2GE effectively attenuated the pro-inflammatory mediators by decreasing the secretion of nitric oxide and cytokines (TNF-α and IL-6) to a greater extent than curcumin due to an increase in cellular uptake. Altogether, the newly developed acid-stable CUR-2GE prodrug is a potential pre-clinical and clinical candidate for further evaluation on neuroprotective and anti-neuroinflammatory effects.


Curcumin , Prodrugs , Aminobutyrates , Carbamates , Curcumin/chemistry , Curcumin/pharmacology , Esters/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , gamma-Aminobutyric Acid
12.
J Ethnopharmacol ; 290: 115088, 2022 May 23.
Article En | MEDLINE | ID: mdl-35149131

ETHNOPHARMACOLOGICAL RELEVANCE: Ben-Cha-Moon-Yai (BMY) remedy used in Thai traditional medicine as an anti-inflammatory, analgesic, and antipyretic agent compromises five herbal root extracts of equal weights: Aegle marmelos (L.) Corrêa (AM), Oroxylum indicum (L.) Kurz (OI), Dimocarpus longan Lour. (DL), Dolichandrone serrulata (Wall. ex DC.) Seem. (DS), and Walsura trichostemon Miq. (WT). AIM OF THE STUDY: To assess the anti-nociceptive and anti-inflammatory effects of the root extracts of all five species of BMY in experimental animal (mouse) models to ensure the rational use of herbal products in Thai traditional medicine. MATERIALS AND METHODS: Root extracts prepared by ethanol and water extraction were used for the biological assays in animal models at five dose levels: 25, 50,100,200 & 400 mg/kg. The anti-nociceptive activity was evaluated based on hot-plate latency, duration of paw licking induced by formalin, and abdominal writhing induced by acetic acid. Carrageenan- and prostaglandin-induced paw oedema models were used to determine the anti-inflammatory activity. RESULTS: The oral administration of AM, DS and WT root extracts displayed significant analgesic effects in the hot-plate test, both phases (early and late) of formalin test and acetic-acid induced writhing test at different dose levels. OI and DL only produced significant analgesia in the late phase of the formalin test and writhing test. The pretreatment of animals with the non-selective opioid receptor antagonist naloxone, reverse AM, DS and WT induced-antinociceptive activity. In both carrageenan and prostaglandin-induced paw oedema tests, all five herbal plant root extracts significantly reduced paw oedema at 3 h or more at different dose levels. Rotarod test results showed no effects of five herbal plant root extracts on the balance and the motor coordination at the highest dose level evaluated (400 mg/kg). CONCLUSION: The root extracts of AM, DS, and WT possess both central and peripheral anti-nociceptive properties, while OI and DL possess only peripheral analgesic properties. All five root extracts own anti-inflammatory properties, which might be due to their activity on the prostaglandin system. Altogether these findings ensure the rational use of BMY remedy in Thai traditional medicine.


Anti-Inflammatory Agents/pharmacology , Nociception/drug effects , Opioid Peptides/drug effects , Plant Extracts/pharmacology , Plant Roots , Prostaglandins/metabolism , Aegle , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Indomethacin/pharmacology , Male , Medicine, East Asian Traditional , Mice , Mice, Inbred ICR , Morphine/pharmacology , Naloxone/pharmacology , Pain Measurement , Plant Preparations/pharmacology , Sapindaceae
13.
PLoS One ; 16(8): e0256706, 2021.
Article En | MEDLINE | ID: mdl-34449819

The use of endotoxin, such as lipopolysaccharide (LPS) as a model of sickness behavior, has attracted recent attention. To objectively investigate sickness behavior along with its pain-like behaviors in LPS-treated mice, the behavioral measurement requires accurate methods, which reflects clinical relevance. While reflexive pain response tests have been used for decades for pain assessment, its accuracy and clinical relevance remain problematic. Hence, we used automated home-cage monitoring LABORAS to evaluate spontaneous locomotive behaviors in LPS-induced mice. LPS-treated mice displayed sickness behaviors including pain-like behaviors in automated home-cage monitoring characterized by decreased mobile behaviors (climbing, locomotion, rearing) and increased immobility compared to that of the control group in both short- and long-term locomotive assessments. Here, in short-term measurement, both in the open-field test and automated home-cage monitoring, mice demonstrated impaired locomotive behaviors. We also assessed 24 h long-term locomotor activity in the home-cage system, which profiled the diurnal behaviors of LPS-stimulated mice. The results demonstrated significant behavioral impairment in LPS-stimulated mice compared to the control mice in both light and dark phases. However, the difference is more evident in the dark phase compared to the light phase owing to the nocturnal activity of mice. In addition, the administration of indomethacin as a pharmacological intervention improved sickness behaviors in the open-field test as well as automated home-cage monitoring, confirming that automated home-cage monitoring could be potentially useful in pharmacological screening. Together, our results demonstrate that automated home-cage monitoring could be a feasible alternative to conventional methods, such as the open-field test and combining several behavioral assessments may provide a better understanding of sickness behavior and pain-like behaviors in LPS-treated mice.


Anxiety/diagnosis , Illness Behavior/physiology , Monitoring, Physiologic , Pain/diagnosis , Animals , Anxiety/diagnostic imaging , Anxiety/physiopathology , Behavior, Animal/physiology , Disease Models, Animal , Humans , Illness Behavior/drug effects , Lipopolysaccharides/toxicity , Locomotion/physiology , Mice , Motor Activity/physiology , Pain/diagnostic imaging , Pain/physiopathology , Pain Measurement/methods
14.
Pharmaceuticals (Basel) ; 14(8)2021 Aug 07.
Article En | MEDLINE | ID: mdl-34451874

Chronic pain is a persistent and unremitting condition that has immense effects on patients' quality of life. Studies have shown that neuroinflammation is associated with the induction and progression of chronic pain. The activation of microglia and astrocytes is the major hallmark of spinal neuroinflammation leading to neuronal excitability in the projection neurons. Excessive activation of microglia and astrocytes is one of the major contributing factors to the exacerbation of pain. However, the current chronic pain treatments, mainly by targeting the neuronal cells, remain ineffective and unable to meet the patients' needs. Curcumin, a natural plant product found in the Curcuma genus, improves chronic pain by diminishing the release of inflammatory mediators from the spinal glia. This review details the role of curcumin in microglia and astrocytes both in vitro and in vivo and how it improves pain. We also describe the mechanism of curcumin by highlighting the major glia-mediated cascades in pain. Moreover, the role of curcumin on inflammasome and epigenetic regulation is discussed. Furthermore, we discuss the strategies used to improve the efficacy of curcumin. This review illustrates that curcumin modulating microglia and astrocytes could assure the treatment of chronic pain by suppressing spinal neuroinflammation.

15.
Sci Rep ; 11(1): 12240, 2021 06 10.
Article En | MEDLINE | ID: mdl-34112846

The failure to develop analgesic drugs is attributed not only to the complex and diverse pathophysiology of pain in humans but also to the poor experimental design and poor preclinical assessment of pain. Although considerable efforts have been devoted to overcoming the relevant problems, many features of the behavioral pain assessment remain to be characterized. For example, a decreased locomotor activity as a common presentation of pain-like behavior has yet to be described. Studies on mice experimentally induced with carrageenan have provided opportunities to explore pain-related behaviors in automated home-cage monitoring. Through this approach, the locomotor activities of mice with carrageenan-induced inflammatory pain can be precisely and objectively captured. Here, we found that the mobile behaviors of mice reduced, and their immobility increased, indicating that carrageenan induction in mice caused a significant decrease in locomotor activity. These non-reflexive pain behaviors were strongly correlated with the reflexive pain behaviors measured via von Frey and plantar tests. Furthermore, the pharmacological intervention using indomethacin improved the locomotor activity of mice with carrageenan-induced pain. Thus, the analysis of the locomotor activity in automated home-cage monitoring is useful for studying the behavioral analgesia and the pharmacological screening of analgesic drugs. The combined evaluation of reflexive and non-reflexive pain behaviors enhances the translational utility of preclinical pain research in rodents.


Behavior, Animal , Inflammation/complications , Instinct , Pain Measurement , Pain/etiology , Animals , Carrageenan , Disease Models, Animal , Mice , Motor Activity , Pain/diagnosis , Temperature
16.
Eur J Pharmacol ; 899: 174008, 2021 May 15.
Article En | MEDLINE | ID: mdl-33705800

Neuropathic pain is a debilitating chronic pain condition, and its treatment remains a clinical challenge. Curcumin, a naturally occurring phenolic compound, possesses diverse biological and pharmacological effects but has not yet been approved as a drug due to its low bioavailability. In order to overcome this limitation, we synthesized a potential ester prodrug of curcumin, curcumin diethyl diglutarate (CurDDG). In this study, we evaluated the pharmacological advantages of CurDDG over curcumin in a mouse model of chronic constriction injury (CCI), and the anti-inflammatory effect of CurDDG in LPS-induced RAW 264.7 macrophage cells was accessed to clarify the underline mechanism. Mice were treated with various oral doses of curcumin (25, 50, 100 and 200 mg/kg/day, daily for 14 days) or equimolar doses of CurDDG. CurDDG at all doses tested significantly attenuated CCI-induced thermal hyperalgesia and mechanical allodynia compared with the CCI-control group. CurDDG at 25, 50 and 100 mg/kg demonstrated significantly greater efficacy on both mechanical and thermal hypersensitivities compared to that of curcumin. The effect of CurDDG correlated well with the inhibition of TNF-α and IL-6 levels in both the sciatic nerve and the spinal cord, as compared to its respective control groups. Similarly, in the in vitro study, CurDDG significantly reduced the LPS-induced expression of TNF-α and IL-6. Moreover, CurDDG significantly decreased COX-2 and iNOS levels and attenuated p38, JNK, and ERK1/2 phosphorylation as compared to the curcumin-treated cells. Altogether, this study demonstrated the improved pharmacological effects of curcumin by its diglutarate conjugate, CurDDG.


Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Curcumin/analogs & derivatives , Curcumin/pharmacology , Glutarates/pharmacology , Hyperalgesia/prevention & control , Pain Threshold/drug effects , Prodrugs/pharmacology , Sciatic Nerve/drug effects , Sciatica/prevention & control , Spinal Cord/drug effects , Animals , Behavior, Animal/drug effects , Cyclooxygenase 2/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred ICR , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , RAW 264.7 Cells , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology , Sciatica/metabolism , Sciatica/physiopathology , Signal Transduction , Spinal Cord/metabolism , Spinal Cord/physiopathology , Succinates , Tumor Necrosis Factor-alpha/metabolism
17.
Molecules ; 25(18)2020 Sep 11.
Article En | MEDLINE | ID: mdl-32933086

Analgesic drugs in a combination-form can achieve greater efficacy with lesser side effects compared to either drug alone. The combination of drugs acting at different targets or mechanisms of action has been recognized as an alternative approach for achieving optimal analgesia. In this study, the analgesic effects of pregabalin (30, 60, 100, 200 mg/kg), curcumin (15, 30, 60, 100, 120 mg/kg), and 1:1 fixed-dose ratio of the pregabalin-curcumin combination were assessed using two acute nociceptive pain models, the acetic acid-induced writhing and tail-flick tests in mice. The pregabalin-curcumin combination produced a dose-dependent decrease in mean of writhes and an increase in the percentage of antinociception by the acetic acid-induced writhing test. In the tail-flick test, the combination also showed an improvement in antinociception indicated by the tail-flick latency, % antinociception, and area under the curve (AUC). Isobolographic analysis of interactions demonstrated a significant synergistic interaction effect between pregabalin and curcumin in both acute nociceptive pain models with the experimental ED50 below the predicted additive line and the combination index < 1. These findings demonstrate that the combination of pregabalin and curcumin exhibits a synergistic interaction in mouse models of acute nociceptive pain.


Analgesics/administration & dosage , Curcumin/administration & dosage , Nociception/drug effects , Pregabalin/administration & dosage , Acetic Acid/chemistry , Administration, Oral , Animals , Area Under Curve , Behavior , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Male , Mice , Mice, Inbred ICR , Pain Management , Pain Measurement
18.
Pharmaceuticals (Basel) ; 13(9)2020 Aug 27.
Article En | MEDLINE | ID: mdl-32867013

The drug treatment for neuropathic pain remains a challenge due to poor efficacy and patient satisfaction. Curcumin has been reported to alleviate neuropathic pain, but its clinical application is hindered by its low solubility and poor oral bioavailability. Curcumin diglutaric acid (CurDG) is a curcumin prodrug with improved water solubility and in vivo antinociceptive effects. In this study, we investigated the anti-inflammatory mechanisms underlying the analgesic effect of CurDG in the chronic constriction injury (CCI)-induced neuropathy mouse model. Repeated oral administration of CurDG at a low dose equivalent to 25 mg/kg/day produced a significant analgesic effect in this model, both anti-allodynic activity and anti-hyperalgesic activity appearing at day 3 and persisting until day 14 post-CCI surgery (p < 0.001) while having no significant effect on the motor performance. Moreover, the repeated administration of CurDG diminished the increased levels of the pro-inflammatory cytokines: TNF-α and IL-6 in the sciatic nerve and the spinal cord at the lowest tested dose (equimolar to 25 mg/kg curcumin). This study provided pre-clinical evidence to substantiate the potential of pursuing the development of CurDG as an analgesic agent for the treatment of neuropathic pain.

19.
Int J Mol Sci ; 21(16)2020 Aug 09.
Article En | MEDLINE | ID: mdl-32784830

Curcumin diglutaric acid (CurDG), an ester prodrug of curcumin, has the potential to be developed as an anti-inflammatory agent due to its improved solubility and stability. In this study, the anti-inflammatory effects of CurDG were evaluated. The effects of CurDG on inflammatory mediators were evaluated in LPS-stimulated RAW 264.7 macrophage cells. CurDG reduced the increased levels of NO, IL-6, and TNF- α, as well as iNOS and COX-2 expression in cells to a greater extent than those of curcumin, along with the potent inhibition of MAPK (ERK1/2, JNK, and p38) activity. The anti-inflammatory effects were assessed in vivo by employing a carrageenan-induced mouse paw edema model. Oral administration of CurDG demonstrated significant anti-inflammatory effects in a dose-dependent manner in mice. The effects were significantly higher compared to those of curcumin at the corresponding doses (p < 0.05). Moreover, 25 mg/kg curcumin did not exert a significant anti-inflammatory effect for the overall time course as indicated by the area under the curve data, while the equimolar dose of CurDG produced significant anti-inflammatory effects comparable with 50, 100, and 200 mg/kg curcumin (p < 0.05). Similarly, CurDG significantly reduced the proinflammatory cytokine expression in paw edema tissues compared to curcumin (p < 0.05). These results provide the first experimental evidence for CurDG as a promising anti-inflammatory agent.


Anti-Inflammatory Agents/pharmacology , Curcumin/pharmacology , Esters/pharmacology , Prodrugs/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Carrageenan , Cell Survival/drug effects , Curcumin/chemistry , Cytokines/metabolism , Edema/chemically induced , Edema/metabolism , Edema/prevention & control , Hindlimb , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred ICR , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Prodrugs/chemistry , RAW 264.7 Cells
20.
Biosci Biotechnol Biochem ; 82(8): 1301-1308, 2018 Aug.
Article En | MEDLINE | ID: mdl-29678124

In this work, a curcumin-diglutaric acid (CurDG) prodrug was synthesized by conjugation of curcumin with glutaric acid via an ester linkage. The water solubility, partition coefficient, release characteristics, and antinociceptive activity of CurDG were compared to those of curcumin. The aqueous solubility of CurDG (7.48 µg/mL) is significantly greater than that of curcumin (0.068 µg/mL). A study in human plasma showed that the CurDG completely releases curcumin within 2 h, suggesting the ability of CurDG to serve as a prodrug of curcumin. A hot plate test in mice showed the highest antinociceptive effect dose of curcumin at 200 mg/kg p.o., whereas CurDG showed the same effect at an effective dose of 100 mg/kg p.o., indicating that CurDG significantly enhanced the antinociceptive effect compared to curcumin. The enhanced antinociceptive effect of CurDG may be due to improved water solubility and increased oral bioavailability compared to curcumin.


Analgesics/chemistry , Analgesics/pharmacology , Curcumin/chemistry , Curcumin/pharmacology , Glutarates/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Administration, Oral , Animals , Biological Availability , Curcumin/pharmacokinetics , Dose-Response Relationship, Drug , Humans , Male , Mice, Inbred ICR , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Solubility , Water
...