1
|
Khawaja G, El-Orfali Y. Silibinin's Effects against Methotrexate-Induced Hepatotoxicity in Adjuvant-Induced Arthritis Rat Model. Pharmaceuticals (Basel) 2024; 17:431. [PMID: 38675395 PMCID: PMC11054686 DOI: 10.3390/ph17040431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 04/28/2024] Open
Abstract
Methotrexate (MTX) is the first drug of choice to treat several diseases, including rheumatoid arthritis. However, its administration is accompanied by severe side effects, most commonly hepatotoxicity. Hence, alternative therapies with a lower toxicity and fewer side effects are needed. This study aimed to investigate the antioxidant and hepatoprotective effects of silibinin (SIL, natural agent) against MTX-induced hepatotoxicity in an adjuvant-induced arthritis (AIA) rat model. Arthritic rats were treated with SIL (100 mg/kg) and/or methotrexate (2 mg/kg). Non-arthritic rats, arthritic untreated rats, and arthritic rats who received the vehicle were followed in parallel. SIL alleviated the systemic consequences of arthritis by restoring lost weight, decreasing the erythrocyte sedimentation rate, and ameliorating joint damage, which was evident both micro- and macroscopically. Additionally, SIL prevented the histopathological alterations in the liver and significantly reduced the liver damage caused by MTX and AIA, as shown by a decrease in the markers of liver damage (ALT and AST). Furthermore, SIL relieved the oxidative stress induced by AIA and MTX in liver tissue by decreasing the lipid peroxidation (MDA) levels and enhancing the antioxidant defense system (GSH levels; catalase and superoxide dismutase (SOD) activities). In conclusion, our results suggest that SIL is a potent antioxidant and hepatoprotective agent in arthritic rats. It markedly attenuated the progression and severity of the arthritic disease and eased the oxidative stress in liver tissue by improving the pro-oxidant/antioxidant balance.
Collapse
Affiliation(s)
- Ghada Khawaja
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut 11-5020, Lebanon;
| | - Youmna El-Orfali
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut 11-5020, Lebanon;
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 11-0236, Lebanon
| |
Collapse
|
2
|
Parthasarathy M, Prince SE. Andrographis paniculata (Burm.f.) Nees Alleviates Methotrexate-Induced Hepatotoxicity in Wistar Albino Rats. Life (Basel) 2023; 13:life13051173. [PMID: 37240818 DOI: 10.3390/life13051173] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
Andrographis paniculata is a herbal plant used in traditional medicinal approaches to treat various ailments and diseases. Methotrexate (MTX) is a clinically used immunosuppressant and anticancer drug. One of the increasing concerns with MTX use is liver toxicity. The aim of this study is to investigate the potential effect of aqueous leaf extract of Andrographis paniculata against methotrexate-induced hepatotoxicity. Wistar albino rats were grouped into five groups, and the drugs were administered. MTX (20 mg/kg b.w.) was intraperitoneally injected into rats on the ninth day alone. Aqueous leaf extract of Andrographis paniculata (500 mg/kg b.w./day) was orally administered for 10 days. We confirmed the beneficial effect of aqueous extracts of Andrographis paniculata on restoring the hepatic enzyme markers, lipid profile, antioxidant level, anti-inflammatory marker (IL-10), anti-apoptosis (bcl-2), significant suppression of inflammatory cytokines (TNF-α, and IL-6), apoptosis marker (caspase 3) and cellular tissue damage caused by MTX. Overall, we revealed that Andrographis paniculata reduces critical aspects of oxidative stress, inflammatory processes, and apoptosis, thus protecting against methotrexate-induced hepatotoxicity.
Collapse
Affiliation(s)
- Manisha Parthasarathy
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Sabina Evan Prince
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| |
Collapse
|
3
|
Concerted regulation of OPG/RANKL/ NF‑κB/MMP-13 trajectories contribute to ameliorative capability of prodigiosin and/or low dose γ-radiation against adjuvant- induced arthritis in rats. Int Immunopharmacol 2022; 111:109068. [PMID: 35944459 DOI: 10.1016/j.intimp.2022.109068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prodigiosin (PDG) is a microbial red dye with antioxidant and anti-inflammatory properties, although its effect on rheumatoid arthritis (RA) remains uncertain. Also, multiple doses of low dose γ- radiation (LDR) have been observed to be as a successful intervention for RA. Thus, the purpose of this study was to investigate the ameliorative potential of PDG and/or LDR on adjuvant-induced arthritis (AIA) in rats. METHODS The anti-inflammatory and anti-arthritic effects of PDG and/or LDR were examined in vitro and in vivo, respectively. In the AIA model, the arthritic indexes, paw swelling degrees, body weight gain, and histopathological assessment in AIA rats were assayed. The impact of PDG (200 µg/kg; p.o) and/or LDR (0.5 Gy) on the levels of pro- and anti-inflammatory cytokines (IL-1β, TNF-α, IL-6, IL-18, IL-17A, and IL-10) as well as the regulation of osteoprotegrin (OPG)/ receptor activator of nuclear factor κB ligand (RANKL)/ nuclear factor-κB (NF-κB)/MMP-13 pathways was determined. Methotrexate (MTX; 0.05 mg/kg; twice/week, i.p) was administered concurrently as a standard anti-arthritic drug. RESULTS PDG and/or LDR markedly diminished the arthritic indexes, paw edema, weigh loss in AIA rats, alleviated the pathological alterations in joints, reduced the levels of pro-inflammatory cytokines IL-1β, TNF-α, IL-6, IL-18, IL-17A, and RANKL in serum and synovial tissues, while increasing anti-inflammatory cytokines IL-10 and OPG levels. Moreover, PDG and/or LDR down-regulated the expression of RANKL, NF-κBp65, MMP13, caspase-3, and decreased the RANKL/OPG ratio, whereas OPG and collagen II were enhanced in synovial tissues. CONCLUSION PDG and/or LDR exhibited obvious anti-RA activity on AIA.
Collapse
|
4
|
Kucharská J, Poništ S, Vančová O, Gvozdjáková A, Uličná O, Slovák L, Taghdisiesfejir M, Bauerová K. Treatment with coenzyme Q10, omega-3-polyunsaturated fatty acids and their combination improved bioenergetics and levels of coenzyme Q9 and Q10 in skeletal muscle mitochondria in experimental model of arthritis. Physiol Res 2021; 70:723-733. [PMID: 34505525 PMCID: PMC8820522 DOI: 10.33549/physiolres.934664] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/10/2021] [Indexed: 01/02/2023] Open
Abstract
Rheumatoid arthritis (RA) and its animal model adjuvant arthritis (AA) are inflammatory diseases characterized by chronic inflammation, systemic oxidative stress and disturbed mitochondrial bioenergetics of skeletal muscle. The present study aimed to evaluate the effects of coenzyme Q10 - CoQ10 (100 mg/kg b.w.), omega-3-polyunsaturated fatty acids - omega-3-PUFA (400 mg/kg b.w.) and their combined treatment in AA on impaired skeletal muscle mitochondrial bioenergetics, inflammation and changes in levels CoQ9 and CoQ10 in plasma. Markers of inflammation (C-reactive protein, monocyte-chemotactic protein-1), antioxidant capacity of plasma, respiratory chain parameters of skeletal muscle mitochondria and concentrations of CoQ9 and CoQ10 in plasma and in muscle tissue were estimated. Treatment of the arthritic rats with CoQ10, omega-3-PUFA alone and in combination partially reduced markers of inflammation and increased antioxidant capacity of plasma, significantly increased concentrations of coenzyme Q in mitochondria and improved mitochondrial function in the skeletal muscle. Combined treatment has similar effect on the mitochondrial function as monotherapies; however, it has affected inflammation and antioxidant status more intensively than monotherapies. Long-term supplementary administration of coenzyme Q10 and omega-3-PUFA and especially their combination is able to restore the impaired mitochondrial bioenergetics and antioxidant status in AA.
Collapse
Affiliation(s)
- J Kucharská
- Pharmacobiochemical Laboratory of Third Department of Internal Medicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic. Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Kour G, Haq SA, Bajaj BK, Gupta PN, Ahmed Z. Phytochemical add-on therapy to DMARDs therapy in rheumatoid arthritis: In vitro and in vivo bases, clinical evidence and future trends. Pharmacol Res 2021; 169:105618. [PMID: 33878447 DOI: 10.1016/j.phrs.2021.105618] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/25/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023]
Abstract
The use of biologically active compounds derived from plants i.e. phytochemicals, have been known for ages for their pharmacological activities in the treatment of autoimmune disorders like rheumatoid arthritis (RA). Besides enormous scientific evidence, the therapeutic potential of phytochemicals is often undervalued. The treatment in RA involves the use of synthetic and biological disease modifying anti-rheumatic drugs (DMARDs). However, the long-term treatment in RA is associated with the risk of gastrointestinal, liver, pulmonary and renal toxicities and serious infections including latent tuberculosis, pneumococcus influenza, herpes zoster and hepatitis. These adverse effects sometimes lead to discontinuation of the therapy. A relatively new vision based on the combination of DMARDs with phytochemicals exhibiting anti-inflammatory, anti-arthritic, anti-oxidant, hepatoprotective and nephroprotective properties for the treatment of RA has achieved substantial importance in the last decade. From this perspective, the present review focuses on the combination of DMARDs (primarily MTX) with phytochemicals that have shown synergistic therapeutic effects while decreasing the toxic repercussions of current RA therapy. The review covers recent evidences of such combination studies that have shown promising results both in experimental arthritic models and clinical arthritis. Few of the combinations including resveratrol, sinomenine, coenzyme Q10 exhibited considerable interest because of their efficacy as an adjuvant to the MTX/standard DMARDs therapy in clinical trials. Besides giving an overview of such combination studies the review also critically discusses the limitations with the use of phytochemicals (e.g. solubility, permeability and bioavailability) compromising their clinical application. Additionally, it stresses upon the need of novel delivery systems and pharmaceutical technologies to increase the therapeutic efficacy of the combination therapy. Overall, the review unveils the potential of phytochemicals in combination with DMARDs with increased tolerability and superior efficacy in further refining the future of the RA therapy.
Collapse
Affiliation(s)
- Gurleen Kour
- Inflammation Pharmacology Division, CSIR, Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, J&K, India; School of Biotechnology, University of Jammu, Baba Saheb Ambedkar Road, Jammu Tawi, 180006 J&K, India
| | - Syed Assim Haq
- Formulation & Drug Delivery Division, CSIR, Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, J&K, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Bijender Kumar Bajaj
- School of Biotechnology, University of Jammu, Baba Saheb Ambedkar Road, Jammu Tawi, 180006 J&K, India
| | - Prem N Gupta
- Formulation & Drug Delivery Division, CSIR, Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, J&K, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Zabeer Ahmed
- Inflammation Pharmacology Division, CSIR, Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, J&K, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
6
|
López-Pedrera C, Villalba JM, Patiño-Trives AM, Luque-Tévar M, Barbarroja N, Aguirre MÁ, Escudero-Contreras A, Pérez-Sánchez C. Therapeutic Potential and Immunomodulatory Role of Coenzyme Q 10 and Its Analogues in Systemic Autoimmune Diseases. Antioxidants (Basel) 2021; 10:antiox10040600. [PMID: 33924642 PMCID: PMC8069673 DOI: 10.3390/antiox10040600] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/14/2022] Open
Abstract
Coenzyme Q10 (CoQ10) is a mitochondrial electron carrier and a powerful lipophilic antioxidant located in membranes and plasma lipoproteins. CoQ10 is endogenously synthesized and obtained from the diet, which has raised interest in its therapeutic potential against pathologies related to mitochondrial dysfunction and enhanced oxidative stress. Novel formulations of solubilized CoQ10 and the stabilization of reduced CoQ10 (ubiquinol) have improved its bioavailability and efficacy. Synthetic analogues with increased solubility, such as idebenone, or accumulated selectively in mitochondria, such as MitoQ, have also demonstrated promising properties. CoQ10 has shown beneficial effects in autoimmune diseases. Leukocytes from antiphospholipid syndrome (APS) patients exhibit an oxidative perturbation closely related to the prothrombotic status. In vivo ubiquinol supplementation in APS modulated the overexpression of inflammatory and thrombotic risk-markers. Mitochondrial abnormalities also contribute to immune dysregulation and organ damage in systemic lupus erythematosus (SLE). Idebenone and MitoQ improved clinical and immunological features of lupus-like disease in mice. Clinical trials and experimental models have further demonstrated a therapeutic role for CoQ10 in Rheumatoid Arthritis, multiple sclerosis and type 1 diabetes. This review summarizes the effects of CoQ10 and its analogs in modulating processes involved in autoimmune disorders, highlighting the potential of these therapeutic approaches for patients with immune-mediated diseases.
Collapse
Affiliation(s)
- Chary López-Pedrera
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
- Correspondence: ; Tel.: +34-957-213795
| | - José Manuel Villalba
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, 14014 Córdoba, Spain; (J.M.V.); (C.P.-S.)
| | - Alejandra Mª Patiño-Trives
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Maria Luque-Tévar
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Nuria Barbarroja
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Mª Ángeles Aguirre
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Alejandro Escudero-Contreras
- Rheumatology Service, Reina Sofia Hospital/Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain; (A.M.P.-T.); (M.L.-T.); (N.B.); (M.Á.A.); (A.E.-C.)
| | - Carlos Pérez-Sánchez
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, 14014 Córdoba, Spain; (J.M.V.); (C.P.-S.)
| |
Collapse
|
7
|
Protective Role of Coenzyme Q10 in Acute Sepsis-Induced Liver Injury in BALB/c Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2020:7598375. [PMID: 33381582 PMCID: PMC7762638 DOI: 10.1155/2020/7598375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 11/18/2022]
Abstract
Sepsis increases the risk of the liver injury development. According to the research works, coenzyme Q10 exhibits hepatoprotective properties in vivo as well as in vitro. Current work aimed at investigating the protective impacts of coenzyme Q10 against liver injury in septic BALB/c mice. The male BALB/c mice were randomly segregated into 4 groups: the control group, the coenzyme Q10 treatment group, the puncture and cecal ligation group, and the coenzyme Q10+cecal ligation and puncture group. Cecal ligation and puncture was conducted after gavagaging the mice with coenzyme Q10 during two weeks. Following 48 h postcecal ligation and puncture, we estimated hepatic biochemical parameters and histopathological changes in hepatic tissue. We evaluated the expression of factors associated with autophagy, pyroptosis, and inflammation. Findings indicated that coenzyme Q10 decreased the plasma levels in alkaline phosphatase, alanine aminotransferase, and aspartate aminotransferase in the cecal ligation and puncture group. Coenzyme Q10 significantly inhibited the elevation of sequestosome-1, interleukin-1β, oligomerization domain-like receptor 3 and nucleotide-binding, interleukin-6, and tumor necrosis factor-α expression levels; coenzyme Q10 also increased beclin 1 levels. Coenzyme Q10 might be a significant agent in the treatment of liver injury induced by sepsis.
Collapse
|
8
|
Wang W, Azizyan RA, Garro A, Kajava AV, Ventura S. Multifunctional Amyloid Oligomeric Nanoparticles for Specific Cell Targeting and Drug Delivery. Biomacromolecules 2020; 21:4302-4312. [PMID: 32885960 DOI: 10.1021/acs.biomac.0c01103] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Natural selection has endorsed proteins with amazing structures and functionalities that cannot be matched by synthetic means, explaining the exponential interest in developing protein-based materials. Protein self-assembly allows fabricating complex supramolecular structures from relatively simple building blocks, a bottom-up strategy naturally employed by amyloid fibrils. However, the design of amyloid-inspired materials with biological activity is inherently difficult. Here, we exploit a modular procedure to generate functional amyloid nanostructures with tight control of their mesoscopic properties. The soft amyloid core of a yeast prion was fused to dihydrofolate reductase through flexible linkers of different sizes. This enabled us to produce, for the first time, biocompatible protein-only amyloid-like oligomeric nanoparticles with defined dimensions in which the embedded enzyme remained highly active, as assessed by biophysical and enzymatic assays. The modular design allowed one to obtain multifunctional nanoparticles by incorporating the antibody-binding Z-domain to the protein fusion. We show how these assemblies can be exploited for antibody-directed targeting of specific cell types and the localized delivery of methotrexate, resulting in the intracellular uptake of the drug by cancer cells and their death. Overall, the novel protein particles we describe in this work might find applications in areas as diverse as biocatalysis, bioimaging, or targeted therapies.
Collapse
Affiliation(s)
- Weiqiang Wang
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Rafayel A Azizyan
- Centre de Recherche en Biologie cellulaire de Montpellier, UMR 5237 CNRS, Université Montpellier, Montpellier 34090, France.,Institut de Biologie Computationnelle, Université Montpellier, Montpellier 34090, France
| | - Adriana Garro
- Universidad Nacional de San Luis IMASL-CONICET, San Luis D5702, Argentina
| | - Andrey V Kajava
- Centre de Recherche en Biologie cellulaire de Montpellier, UMR 5237 CNRS, Université Montpellier, Montpellier 34090, France.,Institut de Biologie Computationnelle, Université Montpellier, Montpellier 34090, France
| | - Salvador Ventura
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
9
|
Qindeel M, Khan D, Ahmed N, Khan S. Surfactant-Free, Self-Assembled Nanomicelles-Based Transdermal Hydrogel for Safe and Targeted Delivery of Methotrexate against Rheumatoid Arthritis. ACS NANO 2020; 14:4662-4681. [PMID: 32207921 DOI: 10.1021/acsnano.0c00364] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Methotrexate (MTX) is the first line agent for therapy against rheumatoid arthritis (RA); however, orally its efficacy is hampered by poor solubility, less permeability, short plasma half-life, and reduced bioavailability. Meanwhile, parenteral formulations are associated with severe adverse effects. In an attempt to improve the efficacy of MTX, we synthesized polycaprolactone-polyethylene glycol-polycaprolactone (PCL-PEG-PCL) triblock copolymer by a ring-opening copolymerization reaction and used it as a carrier for the fabrication of MTX-loaded nanomicelles. Surfactant-free, self-assembled nanomicelles were prepared by nanoprecipitation technique and optimized through central composite design. The optimized nanomicelles exhibited a size distribution of 31 nm and an encapsulation efficiency of 91%. In vitro, the nanomicelles exhibited hemocompatibility, sustained release, and significantly high uptake in lipopolysaccharide activated macrophages. To facilitate application on the skin, optimized nanomicelles were loaded into a Carbopol 934-based hydrogel with eucalyptus oil as a penetration enhancer. Eucalyptus oil significantly improved the permeation of nanomicelles through the skin (p < 0.001). When the hydrogel was applied on the RA mice model, nanomicelles exhibited preferentially highest accumulation in the inflamed joints than other organs. As compared with the free MTX, MTX nanomicelles significantly improved the pharmacokinetic (4.34-fold greater half-life, 3.68-fold higher AUC0-t, and 3.15-fold higher mean residence time) and pharmacodynamic profile ascertained through low inflammatory cytokines expression, improved oxidation protection, recovered behavioral responses, and radiological analysis. MTX nanomicelles-based hydrogel also significantly reduced the hepatotoxicity and did not activate the immune system. These results suggest that the MTX-loaded nanomicelles-based transdermal hydrogel can prove to be a promising agent against RA.
Collapse
Affiliation(s)
- Maimoona Qindeel
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Dildar Khan
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Naveed Ahmed
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Salman Khan
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| |
Collapse
|
10
|
Mitochondrial Coenzyme Q Protects Sepsis-Induced Acute Lung Injury by Activating PI3K/Akt/GSK-3 β/mTOR Pathway in Rats. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5240898. [PMID: 31815144 PMCID: PMC6878790 DOI: 10.1155/2019/5240898] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 10/10/2019] [Accepted: 10/25/2019] [Indexed: 12/21/2022]
Abstract
The aim of our study was to assess the effects of mitochondrial coenzyme Q (MitoQ) on sepsis-induced acute lung injury (ALI) and investigate its possible mechanisms. The cecal ligation and puncture (CLP) method was used to establish a septic ALI model. Rats were randomly divided into Con group, CLP group, MitoQ group, and MitoQ + LY294002 group. The survival rate of the rats was recorded, and the survival rate curve was plotted. Moreover, the ratio of wet/dry weight (W/D) in lung tissue was measured. The activity of myeloperoxidase (MPO) was measured by using the MPO colorimetric activity assay kit. The levels of high-mobility group box 1 (HMGB1) and interleukin-6 (IL-6), macrophage inflammatory protein 2 (MIP2), and keratinocyte chemoattractant (KC) were analyzed by ELISA. The histopathological changes were measured by HE staining, and the lung injury was scored. TUNEL assay was applied to detect the apoptotic cells in lung tissue. The protein expressions were detected by western blot. MitoQ increased the survival rate and alleviated pulmonary edema in septic ALI rats. In addition, MitoQ inhibited the MPO activity and decreased the levels of HMGB1 and IL-6. After treatment with MitoQ, alveolar wall edema, inflammatory cell infiltration, and red blood cell exudation were relieved. MitoQ inhibited cell apoptosis in lung tissue of septic ALI rats. Meanwhile, MitoQ treatment remarkedly increased the expression of p-Akt, p-GSK-3β, and p-mTOR but decreased Bax, caspase-3, caspase-9, Beclin-1, and LC-3II/LC-3I. The effects of MitoQ were significantly reversed by the PI3K inhibitor (LY294002). Our study demonstrated that MitoQ could protect sepsis-induced acute lung injury by activating the PI3K/Akt/GSK-3β/mTOR pathway in rats.
Collapse
|
11
|
Effects of coenzyme Q10 supplementation on matrix metalloproteinases and DAS-28 in patients with rheumatoid arthritis: a randomized, double-blind, placebo-controlled clinical trial. Clin Rheumatol 2019; 38:3367-3374. [DOI: 10.1007/s10067-019-04723-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/24/2019] [Accepted: 07/30/2019] [Indexed: 12/21/2022]
|
12
|
Mahmoud AR, Ali FEM, Abd-Elhamid TH, Hassanein EHM. Coenzyme Q 10 protects hepatocytes from ischemia reperfusion-induced apoptosis and oxidative stress via regulation of Bax/Bcl-2/PUMA and Nrf-2/FOXO-3/Sirt-1 signaling pathways. Tissue Cell 2019; 60:1-13. [PMID: 31582012 DOI: 10.1016/j.tice.2019.07.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 07/09/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022]
Abstract
Coenzyme Q10 (CoQ10) is a component of the mitochondrial electron transport chain and regarded as a strong anti-oxidant agent. In this study, we focused on the mechanistic insights involved in the hepato-protective effects of CoQ10 against hepatic ischemia reperfusion (IR) injury. Our results revealed that CoQ10 significantly improved hepatic dysfunctions and oxidative stress caused by IR injury. Interestingly, as compared to IR subjected rat, CoQ10 inhibited apoptosis by marked down-regulation of both Bax and PUMA genes while the level of Bcl-2 gene was significantly increased. Moreover, CoQ10 up-regulated PI3K, Akt and mTOR protein expressions while it inhibited the expression of both GSK-3β and β-catenin. Additionally, CoQ10 restored oxidant/antioxidant balance via marked activated Nrf-2 protein as well as up-regulation of both Sirt-1 and FOXO-3 genes. Moreover, CoQ10 strongly inhibited inflammatory response through down-regulation of NF-κB-p65 and decrease both JAK1 and STAT-3 protein expressions with a subsequent modulating circulating inflammatory cytokines. Furthermore, histopathological analysis showed that CoQ10 remarkably ameliorated the histopathological damage induced by IR injury. Taken together, our results suggested and proved that CoQ10 provided a hepato-protection against hepatic IR injury via inhibition of apoptosis, oxidative stress, inflammation and their closed related pathways.
Collapse
Affiliation(s)
- Amany R Mahmoud
- Department of Human Anatomy and Embryology, Faculty of Medicine, Assiut University, Assiut, Egypt; Department of Anatomy, Unaizah College of Medicine, Qassim University, Unaizah Al Qassim Region, Saudi Arabia
| | - Fares E M Ali
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
| | - Tarek Hamdy Abd-Elhamid
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| |
Collapse
|
13
|
Saha R, Roychoudhury S, Kar K, Varghese A, Nandi P, Sharma G, Formicki G, Slama P, Kolesarova A. Coenzyme Q10 ameliorates cadmium induced reproductive toxicity in male rats. Physiol Res 2019; 68:141-145. [DOI: 10.33549/physiolres.934000] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
This study aimed at investigating the protective role of CoQ10 against cadmium (Cd)-induced reproductive toxicity in male rats. Adult male Wistar rats were exposed to an acute dose of Cd (25 mg/kg bwt; Cd group), Cd+CoQ10 (25 mg/kg bwt Cd+10 mg CoQ10; Cd-Q10 group) and distilled water (control) in vivo for 15 consecutive days and semen quality was assessed. A significant reduction was noted in sperm concentration, progressive motility, morphology and DNA integrity in both Cd- and Cd-Q10 groups in comparison to control indicating Cd-induced testicular lipid per oxidation (LPO) and decline in indigenous antioxidant defense system as measured by total antioxidant capacity (TAC) (p<0.05). However, simultaneous co-administration of CoQ10 along with Cd (Cd-Q10 group) was able to improve sperm concentration, motility, progressive motility, morphology, DNA integrity, and testicular TAC as well as lower LPO compared to Cd group (p<0.05). Results indicate that used dose of CoQ10 is capable of moderately ameliorating reproductive toxicity of Cd by improving semen quality and reducing testicular oxidative stress.
Collapse
Affiliation(s)
- R. Saha
- Department of Life Science and Bioinformatics, Assam University, Silchar, India
| | - S. Roychoudhury
- Department of Life Science and Bioinformatics, Assam University, Silchar, India
| | - K. Kar
- Mediland Hospital and Research Centre, Itkhola, Silchar, India
| | | | - P. Nandi
- Department of Environmental Science, University of Calcutta, India
| | - G.D. Sharma
- Atal Bihari Vajpayee University, Bilaspur, India
| | - G. Formicki
- Department of Zoology of Vertebrates, Institution of Biology, Pedagogical University of Cracow, Poland
| | - P. Slama
- Department of Animal Morphology, Physiology and Genetics, Mendel University in Brno, Brno, Czech Republic
| | - A. Kolesarova
- Department of Animal Physiology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovak Republic
| |
Collapse
|
14
|
Hasan H, Ismail H, El-Orfali Y, Khawaja G. Therapeutic benefits of Indole-3-Carbinol in adjuvant-induced arthritis and its protective effect against methotrexate induced-hepatic toxicity. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:337. [PMID: 30567538 PMCID: PMC6299996 DOI: 10.1186/s12906-018-2408-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 12/12/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Rheumatoid arthritis (RA) being an incapacitating disease requires early effective intervention. Considering Methotrexate (MTX)- the first line of treatment for RA- intoxicates the liver; therefore, alternative therapies with similar efficacy yet lower cytotoxicity are desired. Indole-3-Carbinol (I3C) which is found in cruciferous vegetables was examined for its possible therapeutic potentials in comparison with MTX by investigating its anti-inflammatory, anti-arthritic, anti-oxidant, and hepatoprotective potentials in adjuvant-induced arthritis (AIA) rat model. METHODS Arthritis was induced in Sprague Dawley rats by injection of Complete Freund's Adjuvant (CFA). Arthritic rats were treated with I3C and/or MTX. To examine the anti-inflammatory and anti-arthritic effect, the following parameters were assessed: body weight, macroscopic scoring of the hind paw, the level of the pivotal cytokines (TNF-α, IL-6) heavily involved in the pathogenesis, spleen index, and erythrocyte sedimentation rate. At a histological level, the tibiotarsal joint was stained with several specific stains. To assess the hepatoprotective and anti-oxidant effects, several oxidative stress parameters were monitored, and the liver histology was examined. RESULTS Both I3C and MTX attenuated the inflammation that was aggravated by arthritis by downregulating the inflammatory markers and mediators and alleviating the histopathological changes affecting the tibiotarsal joint. I3C attenuated the liver impairment that was initiated by arthritis and MTX treatment. It did so by downregulating the pro-oxidants and up-regulating the anti-oxidant defenses and by reducing the pathological changes affecting the liver. CONCLUSION Our results suggest that I3C is as potent as MTX as an anti-inflammatory and anti-arthritic agent. In addition, I3C does so while protecting the liver from damage as opposed to MTX.
Collapse
Affiliation(s)
- Hiba Hasan
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Debbieh, Lebanon
| | - Hanan Ismail
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Debbieh, Lebanon
| | - Youmna El-Orfali
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Debbieh, Lebanon
| | - Ghada Khawaja
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Debbieh, Lebanon
| |
Collapse
|
15
|
Coenzyme Q10 attenuates lung and liver fibrosis via modulation of autophagy in methotrexate treated rat. Biomed Pharmacother 2018; 109:892-901. [PMID: 30551543 DOI: 10.1016/j.biopha.2018.10.133] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 10/07/2018] [Accepted: 10/21/2018] [Indexed: 12/19/2022] Open
Abstract
The present study was conducted to investigate the potential protective effects of coenzyme Q 10 (CoQ10) administration on methotrexate induced lung and liver fibrosis in rat model, and to explore our hypothesis regarding its possible mechanism of action through reactivation of autophagy pathway. Methotrexate induced fibrosis was achieved by intraperitoneal injections twice a week for 4 weeks. A combined treatment of CoQ10 and methotrexate were used. Blood samples for biochemical analysis, lung and livers tissue for biochemical and histopathological analysis, were investigated. Concomitant treatment of CoQ10 & methotrexate caused improvement in histological picture of the lung and liver tissues, liver function and oxidative stress biomarkers, modulation of autophagy genes [mammalian target of rapamycin (m-TOR), Microtubule-associated proteins 1 A/1B light chain 3 (MAP1LC3B), and Sequestosome 1 ubiquitin-binding protein p62 (p62/SQSTM1)] with simultaneous reduction in High Mobility Group Protein B1 (HMGB1). Based on our results we postulated that CoQ10 up regulates autophagy pathway that could explain its protective properties against lung and liver fibrosis caused by methotrexate treatment in current study rat model.
Collapse
|
16
|
Li F, Li H, Luo S, Ran Y, Xie X, Wang Y, Zheng M, Wang M, Zhao Z, Li X. Evaluation of the effect of andrographolide and methotrexate combined therapy in complete Freund's adjuvant induced arthritis with reduced hepatotoxicity. Biomed Pharmacother 2018; 106:637-645. [PMID: 29990853 DOI: 10.1016/j.biopha.2018.07.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/23/2018] [Accepted: 07/01/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Methotrexate is one of the most widely used disease-modifying anti-rheumatic drugs. The hepatotoxicity of methotrexate resulted in poor compliance with therapy. The current study was designed to analyse the combined therapy of andrographolide (AD) and methotrexate (MTX) for complete Freund's adjuvant (CFA)-induced arthritis, focusing on hepatoprotective effects, oxidative stress and arthritic-related cytokines. METHOD Wistar rats were injected with CFA into the right hind paw. Ten days post-CFA injection, the Wistar rats were administered with 1% CMC-Na solution, methotrexate (2 mg/kg/week), AD (50 mg/kg/d) and combined therapy for 35 days. The anti-arthritic effect was assessed by paw volume, X-ray and serum tumour necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β levels. Serum samples were also analysed for glutamic oxaloacetic transaminases (GOT), serum glutamic pyruvic transaminase (GPT), alkaline phosphatase (AKP) and lactate dehydrogenase (LDH). Liver tissue samples were used to examine the cellular antioxidant defence activities using catalase activity (CAT) and GSH as well as GSH-Px and MDA. Histopathology analysis was conducted to evaluate liver damage. RESULTS AD treatment strengthened the anti-arthritic capacity of MTX. AD and MTX-combined therapy additively reduced the inflammatory symptoms in CFA rats. The combined therapy of AD and MTX showed hepatoprotective effect indicated by an improvement in the serum marker, possibly due to antioxidant action and confirmed by liver histopathological changes. Furthermore, the combined therapy significantly reduced serum TNF-α, IL-6 and IL-1β levels. CONCLUSIONS A combined therapy of AD and methotrexate significantly alleviated MTX-induced hepatocellular injury and strengthened the anti-arthritic effect. Further clinical studies should be done to further verify the possibility of combined its clinical usage.
Collapse
Affiliation(s)
- Fenfen Li
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue, Zhengzhou 450001, Henan Province, PR China; Department of Pathophysiology, College of Basic Medical Sciences, Zhengzhou University, 100 Kexue, Zhengzhou 450001, Henan Province, PR China
| | - He Li
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue, Zhengzhou 450001, Henan Province, PR China; Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan Province, PR China
| | - Shasha Luo
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue, Zhengzhou 450001, Henan Province, PR China
| | - Yantao Ran
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue, Zhengzhou 450001, Henan Province, PR China
| | - Xiaoqian Xie
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue, Zhengzhou 450001, Henan Province, PR China
| | - Yale Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue, Zhengzhou 450001, Henan Province, PR China
| | - Ming Zheng
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue, Zhengzhou 450001, Henan Province, PR China
| | - Mengzhen Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue, Zhengzhou 450001, Henan Province, PR China
| | - Zeyue Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue, Zhengzhou 450001, Henan Province, PR China
| | - Xiaotian Li
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue, Zhengzhou 450001, Henan Province, PR China.
| |
Collapse
|
17
|
De S, Kundu S, Chatterjee U, Chattopadhyay S, Chatterjee M. Allylpyrocatechol attenuates methotrexate-induced hepatotoxicity in a collagen-induced model of arthritis. Free Radic Res 2018; 52:698-711. [DOI: 10.1080/10715762.2018.1466391] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Soumita De
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Sunanda Kundu
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Uttara Chatterjee
- Department of Pathology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | | | - Mitali Chatterjee
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| |
Collapse
|
18
|
Paunović MG, Matić MM, Ognjanović BI, Saičić ZS. Antioxidative and haematoprotective activity of coenzyme Q10 and vitamin E against cadmium-induced oxidative stress in Wistar rats. Toxicol Ind Health 2017; 33:746-756. [PMID: 28950793 DOI: 10.1177/0748233717725480] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cadmium (Cd) is a major environmental pollutant, which exerts adverse effects mainly by inducing oxidative stress. Coenzyme Q10 (CoQ10) and vitamin E (VE), naturally occurring antioxidants, improve health condition by inactivating free radicals and enhancing antioxidative defence. The aim of our study was to investigate the protective role of CoQ10 and/or VE pretreatment against Cd-induced haematotoxicity. Wistar albino rats were intramuscularly injected with CoQ10 (20 mg/kg b.w.) and/or VE (20 IU/kg b.w.) or with saline (control group). After 24 h, Cd was injected intraperitoneally (0.4 mg/kg b.w.) and 1 day after, animals were sacrificed. Acute Cd intoxication caused significant changes in haematological and biochemical parameters and altered the glutathione cycle, leading to the formation of lipid peroxidation, while the concentrations and activities of antioxidants (vitamins C and E, superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase) were decreased. CoQ10 and/or VE significantly maintained these values to near-normal levels, afforded additional protection by reducing lipid peroxidation and improved the levels of antioxidants in the blood. Plasma CoQ10 and VE levels negatively correlated with oxidative damage parameters while positively correlated with antioxidative defence parameters. Regarding their effects, CoQ10 and VE were in synergistic interaction. The present study suggested that CoQ10 and VE combination may be beneficial in protecting from Cd-induced haematotoxicity and may be used as a preventive against acute Cd intoxication of exposed people.
Collapse
Affiliation(s)
- Milica G Paunović
- 1 University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Kragujevac, Republic of Serbia
| | - Miloš M Matić
- 1 University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Kragujevac, Republic of Serbia
| | - Branka I Ognjanović
- 1 University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Kragujevac, Republic of Serbia
| | - Zorica S Saičić
- 2 University of Belgrade, Institute for Biological Research "Siniša Stanković", Belgrade, Republic of Serbia
| |
Collapse
|
19
|
Ozer EK, Goktas MT, Kilinc I, Pehlivan S, Bariskaner H, Ugurluoglu C, Iskit AB. Coenzyme Q10 improves the survival, mesenteric perfusion, organs and vessel functions in septic rats. Biomed Pharmacother 2017; 91:912-919. [DOI: 10.1016/j.biopha.2017.05.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/02/2017] [Accepted: 05/04/2017] [Indexed: 01/16/2023] Open
|
20
|
Khattab A, Hassanin L, Zaki N. Self-Nanoemulsifying Drug Delivery System of Coenzyme (Q10) with Improved Dissolution, Bioavailability, and Protective Efficiency on Liver Fibrosis. AAPS PharmSciTech 2017; 18:1657-1672. [PMID: 27677262 DOI: 10.1208/s12249-016-0632-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 09/10/2016] [Indexed: 01/21/2023] Open
Abstract
The aim of our investigation is to develop and characterize self-nanoemulsifying drug delivery systems (SNEDDS) of CoQ10 to improve its water solubility, dissolution rate, and bioavailability, and then evaluate its biochemical and physiological effect on liver cirrhosis in rats compared with CoQ10 powder. SNEDDS are isotropic and thermodynamically stable mixture of oil, surfactant, co-surfactant, and drug that form an oil/water nanoemulsion when added to aqueous phases with soft agitation. Upon administration, self-nanoemulsifying system becomes in contact with gastrointestinal fluid and forms o/w nanoemulsion by the aid of gastrointestinal motility. When the nanoemulsion is formed in the gastrointestinal tract, it presents the drug in a solubilized form inside small nano-sized droplets that provide a large surface area for enhancing the drug release and absorption. Solubility of CoQ10 in various oils, surfactants, and co-surfactants were studied to identify the components of SNEDDS; pseudo-ternary phase diagrams were plotted to identify the efficient self-emulsifying regions. CoQ10-loaded SNEDDS were prepared using isopropyl myristate as oil; Cremophor El, Labrasol, or Tween80 as surfactant; and Transcutol as co-surfactant. The amount of CoQ10 in each vehicle was 3%. The formulations that passed thermostability evaluation test were assessed for particle size analysis, morphological characterization, refractive index, zeta potential, viscosity, electroconductivity, drug release profile, as well as ex vivo permeability. Pharmacokinetics and hepatoprotective efficiency of the optimized SNEDDS of CoQ10 compared with CoQ10 suspension were performed. Results showed that all optimized formulae have the ability to form a good and stable nanoemulsion when diluted with water; the mean droplet size of all formulae was in the nanometric range (11.7-13.5 nm) with optimum polydispersity index values (0.2-0.21). All formulae showed negative zeta potential (-11.3 to -17.2), and maximum drug loading efficiency. One hundred percent of CoQ10 was released from most formulae within 30 min. One hundred percent of CoQ10 was permeated from all formulae through 10 h. The pharmacokinetic study in rabbits revealed a significant increase in bioavailability of CoQ10 SNEDDS to 2.1-fold compared with CoQ10 suspension after oral administration. Comparative effect of the optimized formulae on acute liver injury compared with CoQ10 powder was also studied; it was found that all the liver biochemical markers as alanine transferase (ALT), aspartate amino transferase (AST), alkaline phosphatase (ALP), total protein (TP), and albumin were significantly improved at p < 0.05. Also, histochemical and histopthological studies confirm the biochemical results. Our results suggest the potential use of SNEDDS to increase the solubility of liphophilic drug as poorly water-soluble CoQ10 and improve its oral absorption, so it can be more efficient to improve liver damage compared to CoQ10 powder. These results demonstrated that CoQ10 SNEDDS inhibited thioacetamide (TAA)-induced liver fibrosis mainly through suppression of collagen production.
Collapse
|
21
|
Li W, Wu X, Xu X, Wang W, Song S, Liang K, Yang M, Guo L, Zhao Y, Li R. Coenzyme Q10 Suppresses TNF-α-Induced Inflammatory Reaction In Vitro and Attenuates Severity of Dermatitis in Mice. Inflammation 2016; 39:281-289. [PMID: 26341816 DOI: 10.1007/s10753-015-0248-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Anti-oxidant coenzyme Q10 (Co-Q10) is commonly used in clinic. Recently, Co-Q10 was reported to antagonize TNF-α-induced inflammation and play a protective role in various inflammatory conditions. However, its role in dermatitis is unknown. Herein, RAW264.7 macrophage cell line was cultured with stimulation of TNF-α, and administration of Co-Q10 alleviated TNF-α-mediated inflammatory reaction in vitro. Furthermore, oxazolone-induced dermatitis mice model was established, and treatment of Co-Q10 markedly attenuated dermatitis phenotype in this mice model. Moreover, the protective role of Co-Q10 in vitro and in dermatitis was probably due to its repression on NF-κB signaling. Collectively, Co-Q10 may represent a potential molecular target for prevention and treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Xiaojuan Wu
- Department of Pathology and Physiopathology, Shandong University Medical School, 44 Wenhuaxi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Xiangling Xu
- Department of Gynaecology and Obstetrics, Jinan Central Hospital, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Wenhan Wang
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Sijia Song
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Ke Liang
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Min Yang
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Linlin Guo
- Department of Pathology and Physiopathology, Shandong University Medical School, 44 Wenhuaxi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Yunpeng Zhao
- Department of Orthopaedics, Qilu Hospital, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, People's Republic of China.
| | - Ruifeng Li
- Department of Pathology and Physiopathology, Shandong University Medical School, 44 Wenhuaxi Road, Jinan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
22
|
Mustafa HN, El Awdan SA, Hegazy GA, Abdel Jaleel GA. Prophylactic role of coenzyme Q10 and Cynara scolymus L on doxorubicin-induced toxicity in rats: Biochemical and immunohistochemical study. Indian J Pharmacol 2016; 47:649-56. [PMID: 26729958 PMCID: PMC4689020 DOI: 10.4103/0253-7613.169588] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Objective: The study aims to evaluate the protective effects of coenzyme Q10 (CoQ10) and Cynara scolymus L (CS) on doxorubicin (dox)-induced toxicity. Materials and Methods: Sixty male rats were divided into six groups. Group 1 as a control. Group 2 received dox (10 mg/kg) intraperitoneally. Group 3 received CoQ10 (200 mg/kg). Group 4 received CS (500 mg/kg). Group 5 received CoQ10 (200 mg/kg) and dox (10 mg/kg). Group 6 received CS (500 mg/kg) and dox (10 mg/kg). The rats were then evaluated biochemically and immunohistochemically. Results: Dox produced a significant deterioration of hepatic and renal functional parameters. Moreover, an upsurge of oxidative stress and nitrosative stress markers. The expression of alpha-smooth muscle actin (α-SMA) was increased and proliferating cell nuclear antigen (PCNA) expression was decreased. Administration of CoQ10 and CS resulted in a significant improvement of hepatic and renal functional parameters, and an improvement of both α-SMA and PCNA. Conclusion: It is concluded that pretreatment with CoQ10 and CS is associated with up-regulation of favorable protective enzymes and down-regulation of oxidative stress. That can be advised as a supplement to dox-treated patients.
Collapse
Affiliation(s)
- Hesham N Mustafa
- Department of Anatomy, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sally A El Awdan
- Department of Pharmacology, National Research Center, Cairo, Egypt
| | - Gehan A Hegazy
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Biochemistry, National Research Center, Cairo, Egypt
| | | |
Collapse
|
23
|
Elbarbary NS, Ismail EAR, Farahat RK, El-Hamamsy M. ω-3 fatty acids as an adjuvant therapy ameliorates methotrexate-induced hepatotoxicity in children and adolescents with acute lymphoblastic leukemia: A randomized placebo-controlled study. Nutrition 2016; 32:41-7. [DOI: 10.1016/j.nut.2015.06.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/10/2015] [Accepted: 06/18/2015] [Indexed: 02/07/2023]
|
24
|
Abdollahzad H, Aghdashi MA, Asghari Jafarabadi M, Alipour B. Effects of Coenzyme Q10 Supplementation on Inflammatory Cytokines (TNF-α, IL-6) and Oxidative Stress in Rheumatoid Arthritis Patients: A Randomized Controlled Trial. Arch Med Res 2015; 46:527-33. [DOI: 10.1016/j.arcmed.2015.08.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 08/25/2015] [Indexed: 12/26/2022]
|
25
|
Drug Induced Steatohepatitis: An Uncommon Culprit of a Common Disease. BIOMED RESEARCH INTERNATIONAL 2015; 2015:168905. [PMID: 26273591 PMCID: PMC4529891 DOI: 10.1155/2015/168905] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/27/2015] [Accepted: 02/04/2015] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a leading cause of liver disease in developed countries. Its frequency is increasing in the general population mostly due to the widespread occurrence of obesity and the metabolic syndrome. Although drugs and dietary supplements are viewed as a major cause of acute liver injury, drug induced steatosis and steatohepatitis are considered a rare form of drug induced liver injury (DILI). The complex mechanism leading to hepatic steatosis caused by commonly used drugs such as amiodarone, methotrexate, tamoxifen, valproic acid, glucocorticoids, and others is not fully understood. It relates not only to induction of the metabolic syndrome by some drugs but also to their impact on important molecular pathways including increased hepatocytes lipogenesis, decreased secretion of fatty acids, and interruption of mitochondrial β-oxidation as well as altered expression of genes responsible for drug metabolism. Better familiarity with this type of liver injury is important for early recognition of drug hepatotoxicity and crucial for preventing severe forms of liver injury and cirrhosis. Moreover, understanding the mechanisms leading to drug induced hepatic steatosis may provide much needed clues to the mechanism and potential prevention of the more common form of metabolic steatohepatitis.
Collapse
|