1
|
Kaufman MJ, Meloni EG. Xenon gas as a potential treatment for opioid use disorder, alcohol use disorder, and related disorders. Med Gas Res 2025; 15:234-253. [PMID: 39812023 PMCID: PMC11918480 DOI: 10.4103/mgr.medgasres-d-24-00063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/23/2024] [Accepted: 09/26/2024] [Indexed: 01/16/2025] Open
Abstract
Xenon gas is considered to be a safe anesthetic and imaging agent. Research on its other potentially beneficial effects suggests that xenon may have broad efficacy for treating health disorders. A number of reviews on xenon applications have been published, but none have focused on substance use disorders. Accordingly, we review xenon effects and targets relevant to the treatment of substance use disorders, with a focus on opioid use disorder and alcohol use disorder. We report that xenon inhaled at subsedative concentrations inhibits conditioned memory reconsolidation and opioid withdrawal symptoms. We review work by others reporting on the antidepressant, anxiolytic, and analgesic properties of xenon, which could diminish negative affective states and pain. We discuss research supporting the possibility that xenon could prevent analgesic- or stress-induced opioid tolerance and, by so doing could reduce the risk of developing opioid use disorder. The rapid kinetics, favorable safety and side effect profiles, and multitargeting capability of xenon suggest that it could be used as an ambulatory on-demand treatment to rapidly attenuate maladaptive memory, physical and affective withdrawal symptoms, and pain drivers of substance use disorders when they occur. Xenon may also have human immunodeficiency virus and oncology applications because its effects relevant to substance use disorders could be exploited to target human immunodeficiency virus reservoirs, human immunodeficiency virus protein-induced abnormalities, and cancers. Although xenon is expensive, low concentrations exert beneficial effects, and gas separation, recovery, and recycling advancements will lower xenon costs, increasing the economic feasibility of its therapeutic use. More research is needed to better understand the remarkable repertoire of effects of xenon and its potential therapeutic applications.
Collapse
|
2
|
Abbasi E, Mirzaei F, Ghaleiha A, Pourjafar M, Ahmadi M, Mirzajani SS. Effects of cerium oxide nanoparticle on liver oxidative stress and morphological changes in opium withdrawal rats. Toxicol Rep 2025; 14:102025. [PMID: 40290115 PMCID: PMC12032377 DOI: 10.1016/j.toxrep.2025.102025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/08/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Since oxidative stress increased during opioid withdrawal, this study aimed to evaluate the effects of cerium oxide nanoparticles (CeONP) on oxidative stress markers in rat livers. Male Wistar rats were randomly divided into 3 groups, including 1: control rats, 2: withdrawal group, and 3: withdrawal rats received CeONP. Opium administration was administered for 30 days. Then the withdrawal period started and CeONP (0.1 mg/kg) was administrated intravenously for 14 days. After that, the liver was removed, and serum was prepared. The expression of superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT) were measured by Real-time PCR. The amount of malondialdehyde (MDA), total oxidant status (TOS), total antioxidant capacity (TAC), and glutathione levels were determined by the available kit. Blood glucose levels and liver enzymes were measured by using the colorimetric method. A light microscope evaluated liver morphological changes. The liver biomarkers (ALT and AST) and glucose levels significantly increased in the withdrawal group (P < 0.05). Furthermore, the MDA and TOS increased dramatically in the withdrawal group (P < 0.001). However, the levels of GSH and TAC were reduced in the withdrawal group(P < 0.05). The gene expressions and activities of SOD, CAT, and GPx were reduced in the withdrawal group (P < 0.05). However, treatment of withdrawal rats with CeONP normalized all these factors(P < 0.05). CeONP restored histological alterations in the liver. Administration of opium causes an increase in oxidative stress and the activity of liver enzymes. However, treatment of withdrawal rats with CeONP showed potential hepatoprotective effects.
Collapse
Affiliation(s)
- Ebrahim Abbasi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Mirzaei
- Department of Anatomical Sciences, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Ghaleiha
- Behavioral Disorders and Substance Abuse Research Center, Institute of Neuroscience and Mental Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mona Pourjafar
- Department of Molecular Medicine and Genetics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehrdad Ahmadi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Somayeh Mirzajani
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
3
|
Liu G, Li R, Gao J, Lin C, Li H, Peng Y, Wang H, Wang X. Mitigating Cannabidiol's Non-Selective Cytotoxicity via Subcellular Organelle Targeting: Exploring Mitochondrial Targeting Potential. Bioconjug Chem 2025; 36:980-992. [PMID: 40198104 DOI: 10.1021/acs.bioconjchem.5c00012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Cannabidiol (CBD), a phytocannabinoid from Cannabis sativa, is renowned for its nonpsychoactive properties and therapeutic potential. However, its clinical application is limited by nonselective cytotoxicity, affecting microglia, oligodendrocytes, and other cells. To address this, subcellular organelle-targeting strategies were explored to minimize off-target effects and enhance CBD's therapeutic index. Three organelle-specific conjugates targeting mitochondria, endoplasmic reticulum, and lysosomes were synthesized. Among these, the mitochondria-targeting triphenylphosphonium (TPP)-modified CBD conjugates demonstrated reduced cytotoxicity and enhanced anti-inflammatory activity. Further optimization identified a four-carbon ether chain linker (CBD-TPP-C4) that increased antineuroinflammatory activity by 3-fold and reduced cytotoxicity by 1.6-fold, compared to unmodified CBD. CBD-TPP-C4 also elevated mitochondrial ATP levels in vitro, improved mitochondrial morphology and locomotor function in Caenorhabditis elegans, and potentiated morphine analgesia in mice. These findings highlight subcellular targeting as a promising strategy to enhance CBD's safety and efficacy, paving the way for improved therapeutic applications.
Collapse
Affiliation(s)
- Genglian Liu
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Ru Li
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Jingwei Gao
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Cong Lin
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Yinghua Peng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
4
|
Zhuang W, Mun SY, Park WS. Direct effects of antipsychotics on potassium channels. Biochem Biophys Res Commun 2025; 749:151344. [PMID: 39842331 DOI: 10.1016/j.bbrc.2025.151344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Schizophrenia (SCZ) and bipolar disorder (BD) and are severe psychiatric conditions that contribute to disability and increased healthcare costs globally. Although first-, second-, and third-generation antipsychotics are available for treating BD and SCZ, most have various side effects unrelated to their unique functions. Many antipsychotics affect K+ channels (Kv, KCa, Kir, K2P, and other channels), which change the functions of various organs. This review summarizes the biological actions of antipsychotics, including off-target side effects involving K+ channels.
Collapse
Affiliation(s)
- Wenwen Zhuang
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Seo-Yeong Mun
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea.
| |
Collapse
|
5
|
Mansour AMA, Khattab MM, El-Khatib AS, Awaad AK, El-Refaie WM, El-Mezayen NS. Valsartan as a prophylactic treatment against breast cancer development and niche activation: What molecular sequels follow chronic AT-1R blockade? Life Sci 2024; 353:122939. [PMID: 39094905 DOI: 10.1016/j.lfs.2024.122939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/07/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
AIMS Transactivation of insulin-growth-factor-receptor (IGF-1R) by angiotensin-II-type-1-receptor (AT-1R) was only demonstrated in vascular-smooth-muscle cells and has never been tested in breast-cancer (BC). This investigation addressed the impact of chronic AT-1R blockade by valsartan (Val) on possible concurrent AT-1R/IGF-1R signaling inhibition, regressing BC-tumor-microenvironment (TME) cellular components activation, and hindering BC development. MAIN METHODS The effect of different Val doses (10, 20, 40 & 80 mg/kg/day for 490 days) was tested on dimethylbenz(a)anthracene (DMBA)-induced progesterone-promoted-BC in rats. The influence on intratumoral/circulating angiotensin-II (ANG-II) levels and AT-1R/Mas-R immunofluorescent-expression were assessed. The potential AT-1R/IGF-1R crosstalk within TME-BC-stem-cells (BCSCs) and cancer-associated-fibroblasts (CAFs) was evaluated by fluorescently marking these cells and locating the immunofluorescently-stained AT-1R/IGF-1R in them using confocal-laser-microscopy and further quantified by flow cytometry. In addition, the molecular alterations following blocking AT-1R were inspected including determining Src; crucial for IGF-1R transactivation by AT-1R, Notch-1; IGF-IR transcriptional-regulator, and PI3K/Akt &IL-6/STAT expression. Further, the suppression of CSCs' capabilities to maintain pluripotency, stemness features, epithelial-to-mesenchymal-transition (EMT), and angiogenesis was evaluated by assessing NANOG gene, aldehyde-dehydrogenase (ALDH), N-cadherin and vascular-endothelial-growth-factor (VEGF), respectively. Furthermore, the proliferative marker; Ki-67, was detected by immunostaining, and tumors were histologically graded using Elston-Ellis-modified-Scarff-Bloom-Richardson method. KEY FINDINGS Prophylactic Val significantly reduced tumor size, prolonged latency, reduced tumor histopathologic grade, decreased circulating/intratumoral-ANG-II levels, increased Mas-R, and decreased AT1R expression. AT-1R/IGF-1R were co-expressed with a high correlation coefficient on CAFs/BCSCs. Moreover, Val significantly attenuated IGF-1R transactivation and transcriptional regulation via Src and Notch-1 genes' downregulation and reduced Src/IGF-IR-associated PI3K/Akt and IL-6/STAT3 signaling. Further, Val significantly decreased intratumoral NANOG, ALDH, N-cadherin, VEGF, and Ki-67 levels. SIGNIFICANCE Chronic Val administration carries a potential for repurposing as adjuvant or conjunct therapy for patients at high risk for BC.
Collapse
Affiliation(s)
- Amira M A Mansour
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt
| | - Aiman S El-Khatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt
| | - Ashraf K Awaad
- Center of Excellence for Research in Regenerative Medicine and Applications, Faculty of Medicine, Alexandria University, Egypt
| | - Wessam M El-Refaie
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Pharos University in Alexandria, Egypt
| | - Nesrine S El-Mezayen
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt.
| |
Collapse
|
6
|
Abdel-Wahab BA, Zafaar D, Habeeb MS, El-Shoura EAM. Nicorandil mitigates arsenic trioxide-induced lung injury via modulating vital signalling pathways SIRT1/PGC-1α/TFAM, JAK1/STAT3, and miRNA-132 expression. Br J Pharmacol 2024; 181:3215-3231. [PMID: 38741475 DOI: 10.1111/bph.16414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/13/2024] [Accepted: 03/01/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND AND PURPOSE Nicorandil, a selective opener of potassium channels, used to treat angina, has drawn attention for its potential in mitigating lung injury, positioning it as a promising therapeutic approach to treat drug-induced lung toxicity. This study aimed to explore the protective role of nicorandil in arsenic trioxide (ATO)-induced lung injury and to elucidate the underlying mechanistic pathways. EXPERIMENTAL APPROACH We assessed the effects of nicorandil (15 mg·kg-1, p.o.) in a rat model of pulmonary injury induced by ATO (5 mg·kg-1, i.p.). The assessment included oxidative stress biomarkers, inflammatory cytokine levels, and other biomarkers, including sirtuin-1, sirtuin-3, STAT3, TFAM, and JAK in lung tissue. Histological examination using H&E staining and molecular investigations using western blotting and PCR techniques were conducted. KEY RESULTS In our model of lung injury, treatment with nicorandil ameliorated pathological changes as seen with H&E staining, reduced tissue levels of toxicity markers, and exerted significant antioxidant and anti-inflammatory actions. On a molecular level, treatment with nicorandil down-regulated JAK, STAT3, PPARγ, Nrf2, VEGF, p53, and micro-RNA 132 while up-regulating Sirt1, 3, TFAM, AMPK, and ERR-α in lung tissue. CONCLUSIONS AND IMPLICATIONS The results presented here show nicorandil as a significant agent in attenuating lung injury induced by ATO in a rodent model. Nonetheless, further clinical studies are warranted to strengthen these findings.
Collapse
Affiliation(s)
- Basel A Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Dalia Zafaar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University of Technology, and Information, Cairo, Egypt
| | | | - Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
| |
Collapse
|
7
|
Wang Y, Chen X, Qian W, Chen T, Zhang S, Zhang W. Determination of trace morphine and its metabolites in mouse urine using a TpBD functionalized bivalve magnetic nano-adsorbent. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:1748-1755. [PMID: 38437029 DOI: 10.1039/d4ay00080c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
In this study, a new type of covalent organic framework (TpBD) functionalized bivalved magnetic microsphere (TpBD-DS MNS) adsorbent was applied for the enrichment and detection of trace morphine and its metabolites in mouse urine. The main factors affecting the efficiency of magnetic solid phase extraction were optimized, and the optimal MSPE conditions were obtained. Combined with the UPLC-MS/MS technique, a new method for determining trace morphine and its metabolites in urine was established. The detection (LOD) and quantification (LOQ) limits for morphine and its metabolites ranged from 0.16 pg mL-1 to 0.53 pg mL-1 and 0.26 pg mL-1 to 1.25 pg mL-1, respectively. The recovery of the methods ranged from 87.4-97.3%, and the RSD was less than 5%. By employing this methodology, we successfully obtained the temporal change curve of morphine and its metabolites in mouse urine through collection and measurement post intravenous administration of morphine. This approach not only presents a novel means for investigating pharmacokinetics and drug monitoring but also demonstrates significant potential in the fields of forensic toxicology and drug abuse surveillance.
Collapse
Affiliation(s)
- Yuancheng Wang
- College of Chemistry, Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China.
| | - Xin Chen
- College of Chemistry, Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China.
| | - Wenping Qian
- College of Chemistry, Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China.
| | - Tianqi Chen
- School of Ecology and Environment, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Shusheng Zhang
- College of Chemistry, Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China.
- Food Laboratory of Zhongyuan, Flavour Science Research Center of Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China.
| | - Wenfen Zhang
- College of Chemistry, Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China.
- Food Laboratory of Zhongyuan, Flavour Science Research Center of Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China.
| |
Collapse
|
8
|
Tu Y, Han D, Liu Y, Hong D, Chen R. Nicorandil attenuates cognitive impairment after traumatic brain injury via inhibiting oxidative stress and inflammation: Involvement of BDNF and NGF. Brain Behav 2024; 14:e3356. [PMID: 38376046 PMCID: PMC10757892 DOI: 10.1002/brb3.3356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 11/26/2023] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Cognitive impairment is a prevalent adverse consequence of traumatic brain injury (TBI). The neuroprotective effects of nicorandil (N-(2-hydroxyethyl)-nicotinamide nitrate) has been previously documented, yet its protective effects against cognitive dysfunction post-TBI remain unclear. Hence, the present study was aimed to evaluate whether nicorandil attenuates cognitive dysfunction in TBI rats and the underlying mechanism behind this process. METHODS The TBI model was established with a controlled cortical impact (CCI). The effects of nicorandil on cognitive dysfunction of rats with TBI were examined through Novel object recognition (NOR) test, Y-maze test, and Morris water maze (MWM) task. After behavioral tests, hippocampal tissue was collected for Quantitative real-time PCR, Western blot analysis, and Enzyme-linked immunosorbent assay (ELISA) assay. RESULTS We observed that nicorandil administration effectively ameliorates learning and memory impairment in TBI rats. Alongside, nicorandil treatment attenuated oxidative stress in the hippocampus of TBI rats, characterized by the decreased reactive oxygen species generation, malondialdehyde, and protein carbonyls levels, and concurrent promotion of antioxidant-related factors (including superoxide dismutase, glutathione peroxidase, and catalase) activities. Additionally, nicorandil treatment attenuated the inflammatory response in the hippocampus of TBI rat, as evidenced by the upregulated levels of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α), as well as the downregulated level of IL-10. Mechanistically, nicorandil treatment significantly enhanced the mRNA and protein levels of neurotrophic factors, brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) in the hippocampus of TBI rats. CONCLUSION These findings suggest that nicorandil mitigates cognitive impairment after TBI by suppressing oxidative stress and inflammation, potentially through enhancing BDNF and NGF levels.
Collapse
Affiliation(s)
- Yaoyan Tu
- Department of Emergency and Trauma CenterNanchang First HospitalNanchangJiangxiChina
| | - Desen Han
- Department of Emergency and Trauma CenterNanchang First HospitalNanchangJiangxiChina
| | - Yanjun Liu
- Department of Emergency and Trauma CenterNanchang First HospitalNanchangJiangxiChina
| | - Dequan Hong
- Department of Emergency and Trauma CenterNanchang First HospitalNanchangJiangxiChina
| | - Rehua Chen
- Department of Emergency and Trauma CenterNanchang First HospitalNanchangJiangxiChina
| |
Collapse
|
9
|
Xu Y, Dong X, Xu H, Jiao P, Zhao LX, Su G. Nanomaterial-Based Drug Delivery Systems for Pain Treatment and Relief: From the Delivery of a Single Drug to Co-Delivery of Multiple Therapeutics. Pharmaceutics 2023; 15:2309. [PMID: 37765278 PMCID: PMC10537372 DOI: 10.3390/pharmaceutics15092309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The use of nanomaterials in drug delivery systems for pain treatment is becoming increasingly common. This review aims to summarize how nanomaterial-based drug delivery systems can be used to effectively treat and relieve pain, whether via the delivery of a single drug or a combination of multiple therapeutics. By utilizing nanoformulations, the solubility of analgesics can be increased. Meanwhile, controlled drug release and targeted delivery can be realized. These not only improve the pharmacokinetics and biodistribution of analgesics but also lead to improved pain relief effects with fewer side effects. Additionally, combination therapy is frequently applied to anesthesia and analgesia. The co-encapsulation of multiple therapeutics into a single nanoformulation for drug co-delivery has garnered significant interest. Numerous approaches using nanoformulation-based combination therapy have been developed and evaluated for pain management. These methods offer prolonged analgesic effects and reduced administration frequency by harnessing the synergy and co-action of multiple targets. However, it is important to note that these nanomaterial-based pain treatment methods are still in the exploratory stage and require further research to be effectively translated into clinical practice.
Collapse
Affiliation(s)
- Yuhang Xu
- School of Pharmacy, Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Xingpeng Dong
- School of Pharmacy, Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Heming Xu
- School of Pharmacy, Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Peifu Jiao
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan 250200, China
| | - Lin-Xia Zhao
- School of Pharmacy, Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Gaoxing Su
- School of Pharmacy, Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong 226019, China
| |
Collapse
|
10
|
Bedair AF, Wahid A, El-Mezayen NS, El-Yazbi AF, Khalil HA, Hassan NW, Afify EA. Nicorandil/ morphine crosstalk accounts for antinociception and hepatoprotection in hepatic fibrosis in rats: Distinct roles of opioid/cGMP and NO/KATP pathways. Biomed Pharmacother 2023; 165:115068. [PMID: 37392650 DOI: 10.1016/j.biopha.2023.115068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/03/2023] Open
Abstract
Previous report indicated that nicorandil potentiated morphine antinociception and attenuated hepatic injury in liver fibrotic rats. Herein, the underlying mechanisms of nicorandil/morphine interaction were investigated using pharmacological, biochemical, histopathological, and molecular docking studies. Male Wistar rats were injected intraperitoneally (i.p.) with carbon tetrachloride (CCl4, 40%, 2 ml/kg) twice weekly for 5 weeks to induce hepatic fibrosis. Nicorandil (15 mg/kg/day) was administered per os (p.o.) for 14 days in presence of the blockers; glibenclamide (KATP channel blocker, 5 mg/kg, p.o.), L-NG-nitro-arginine methyl ester (L-NAME, nitric oxide synthase inhibitor, 15 mg/kg, p.o.), methylene blue (MB, guanylyl cyclase inhibitor, 2 mg/kg, i.p.) and naltrexone (opioid antagonist, 20 mg/kg, i.p.). At the end of the 5th week, analgesia was evaluated using tail flick and formalin tests along with biochemical determinations of liver function tests, oxidative stress markers and histopathological examination of liver tissues. Naltrexone and MB inhibited the antinociceptive activity of the combination. Furthermore, combined nicorandil/morphine regimen attenuated the release of endogenous peptides. Docking studies revealed a possible interaction of nicorandil on µ, κ and δ opioid receptors. Nicorandil/morphine combination protected against liver damage as evident by decreased liver enzymes, liver index, hyaluronic acid, lipid peroxidation, fibrotic insults, and increased superoxide dismutase activity. Nicorandil/morphine hepatoprotection and antioxidant activity were inhibited by glibenclamide and L-NAME but not by naltrexone or MB. These findings implicate opioid activation/cGMP versus NO/KATP channels in the augmented antinociception, and hepatoprotection, respectively, of the combined therapy and implicate provoked cross talk by nicorandil and morphine on opioid receptors and cGMP signaling pathway. That said, nicorandil/morphine combination provides a potential multitargeted therapy to alleviate pain and preserve liver function.
Collapse
Affiliation(s)
- Asser F Bedair
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Alexandria, Alexandria, Egypt
| | - Ahmed Wahid
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, University of Alexandria, Alexandria, Egypt
| | - Nesrine S El-Mezayen
- Department of Pharmacology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Amira F El-Yazbi
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, University of Alexandria, Alexandria, Egypt
| | - Hadeel A Khalil
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, University of Alexandria, Alexandria, Egypt
| | - Nayera W Hassan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Alexandria, Alexandria, Egypt
| | - Elham A Afify
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Alexandria, Alexandria, Egypt.
| |
Collapse
|
11
|
Zhang S, Zhu P, Yuan J, Cheng K, Xu Q, Chen W, Pan Z, Zheng Y. Non-alcoholic fatty liver disease combined with rheumatoid arthritis exacerbates liver fibrosis by stimulating co-localization of PTRF and TLR4 in rats. Front Pharmacol 2023; 14:1149665. [PMID: 37346294 PMCID: PMC10279862 DOI: 10.3389/fphar.2023.1149665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/25/2023] [Indexed: 06/23/2023] Open
Abstract
Rheumatoid arthritis (RA) has a high prevalence in patients with non-alcoholic fatty liver disease (NAFLD); however, the underlying mechanism is unclear. To address this, our study established a rat model with both NAFLD and RA by feeding a high-fat diet (HFD) and administering intradermal injection of Freund's complete adjuvant (FCA) with bovine type II collagen. Collagen-induced RA (CIA) was confirmed by hind paw swelling and histological examination. The histomorphological characteristics of NAFLD were evaluated by Masson's trichrome and hematoxylin-eosin staining. The development of NAFLD was further evaluated by measuring serum concentrations of triglyceride (TG), total cholesterol (T-CHO), alanine aminotransferase (ALT), aspartate aminotransferase (AST), and lipopolysaccharide (LPS). The results showed that HFD feeding exacerbated secondary inflammation in CIA rats, whereas FCA/bovine type II collagen injection increased serum levels of ALT, AST, TG, T-CHO, and LPS and exacerbated hepatic fibrosis in both normal and NAFLD rats. Interestingly, NAFLD + CIA significantly promoted the expression of PTRF, a caveolae structure protein involved in hepatic lipid metabolism and affecting downstream signaling of Toll-like receptor 4 (TLR4) and PI3K/Akt activation. High resolution confocal microscopy revealed increased PTRF and TLR4 co-localization in hepatic small vessels of NAFLD + CIA rats. AAV9-mediated PTRF knockdown inhibited TLR4 signaling and alleviated hepatic fibrosis in NAFLD + CIA rats. Together, these findings indicate that NAFLD combined with CIA causes synovial injury and enhances non-alcoholic fatty liver fibrosis in rats. PTRF could attenuate the symptoms of NAFLD + CIA likely by affecting TLR4/PTRF co-expression and downstream signaling.
Collapse
Affiliation(s)
| | - Peng Zhu
- School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Jianan Yuan
- School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Kunming Cheng
- School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Qixiang Xu
- School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Wei Chen
- Boster Biological Technology Co., Ltd., Wuhan, China
| | - Zui Pan
- College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, United States
| | - Yongqiu Zheng
- School of Pharmacy, Wannan Medical College, Wuhu, China
| |
Collapse
|