1
|
Zhao XL, Xu LY, Li KD, Tang F, Liu D, Zhang JN, Cao ZJ, Peng C, Ao H. Exploring dried ginger essential oil as a therapeutic strategy for 5-FU-induced mucositis: Gut microbiota and tryptophan metabolite IAA-AHR/IL-22/STAT3 signaling axis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119616. [PMID: 40074099 DOI: 10.1016/j.jep.2025.119616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/01/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 5-Fluorouracil (5-FU) commonly induces severe mucositis, causing pain, inflammation, and gastrointestinal dysfunction, which significantly increases patient morbidity and reduces quality of life. In Ayurveda, Traditional Chinese Medicine, and other ethnopharmacological practices, dried ginger has been widely used to alleviate symptoms such as nausea, vomiting, diarrhea, and inflammation, highlighting its important role in traditional medicine. AIM OF THE STUDY This study explored the potential of dried ginger essential oil (DGEO) in mitigating intestinal epithelial barrier damage in mice with mucositis induced by 5-FU. METHODS The therapeutic effects of DGEO were evaluated by measurements of weight changes, diarrhea scores, ELISA, and H&E. Further investigations included 16S rRNA sequencing, untargeted metabolomics, molecular docking, and HPLC-MS/MS to explore its underlying mechanisms, with validation performed using western blotting and ELISA. RESULTS The results demonstrated that DGEO was effective in alleviating mucositis symptoms. It also improved the gut microbiota, enhanced the biotransformation of tryptophan to indole-3-acetic acid (IAA), and elevated the protein expressions of the AHR, CYP1A1, and p-STAT3, as well as level of IL-22. Moreover, DGEO improved the expressions of tight junction (TJ) proteins and anti-apoptotic proteins, enhancing intestinal barrier integrity. CONCLUSION These findings indicated that DGEO ameliorated 5-FU-induced mucositis by modulating gut microbiota and the tryptophan metabolite IAA-AHR/IL-22/STAT3 signaling axis, providing new insights into its therapeutic applications, particularly its ability to regulate gut microbiota and related signaling pathways.
Collapse
Affiliation(s)
- Xiao-Lan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li-Yue Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke-Di Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing-Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhang-Jing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
2
|
Tirgar P, Vekaria M, Raval K. Pre-clinical Evaluation of Karanjin Against DMBA-Induced Breast Cancer in Female Sprague-Dawley Rats Through Modulation of SMAR1 and CDP/CUx genes. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1825-1839. [PMID: 39177785 DOI: 10.1007/s00210-024-03389-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024]
Abstract
PURPOSE To investigate the chemoprotective potential of karanjin against 7,12-dimethylbenz(α)anthracene (DMBA)-induced breast cancer. METHODOLOGY Thirty-six female rats were utilized for the study. Breast cancer was induced through a subcutaneous injection of 35 mg/kg DMBA. The animals were allocated to six groups. Three groups were allocated for karanjin (50 mg/kg, 100 mg/kg, and 200 mg/kg), and received daily treatment for 20 weeks (including 2 weeks as pre-treatment). Doxorubicin (4 mg/kg) was administered to the standard control group twice a week for 20 weeks. The disease control (DC) and normal control (NC) groups received daily treatment with saline. After the treatment, oxidative stress parameters, biochemical parameters, and inflammatory parameters were estimated. CCAAT-displacement protein/cut homeobox (CUP/Cux) and scaffold/matrix attachment region binding protein 1 (SMAR1) expression levels were measured through gene expression analysis. Immunohistochemical (IHC) analysis was performed to estimate the expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2). RESULTS Tumor growth reduced significantly (P-value < 0.01) in karanjin-treated animals compared to the DC group. Karanjin significantly (P-value < 0.01) regulated the levels of oxidative stress parameters, biochemical parameters, and inflammatory parameters compared to the DC group. Karanjin treatment significantly (P-value < 0.001) regulated the expression levels of SMAR1 and CDP/Cux. A notable reduction in the IHC scores was observed for ER, PR, and HER2 expression in karanjin groups. CONCLUSION Karanjin demonstrated chemoprotective activity against DMBA-induced breast cancer in animals potentially through modulation of SMAR1 and CDP/Cux gene expression and reduction of ER, PR and HER2 expression levels.
Collapse
MESH Headings
- Animals
- Female
- 9,10-Dimethyl-1,2-benzanthracene/toxicity
- Rats, Sprague-Dawley
- Mammary Neoplasms, Experimental/chemically induced
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/prevention & control
- Rats
- Oxidative Stress/drug effects
- Receptor, ErbB-2/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
Collapse
Affiliation(s)
- Pravin Tirgar
- School of Pharmacy, RK University, Rajkot, Gujarat, India
| | | | - Keval Raval
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa-388421, Anand, Gujarat, India.
| |
Collapse
|
3
|
Hou Y, Zhao Y, Liu J, Bao Y, Mosesmanaanye N, Zhao C, Li W, Hong B. Optimization of Docetaxel-Zedoary Turmeric Oil Magnetic Solid Lipid Nanoparticle Preparation by Central Composite Design-Response Surface Methodology. Assay Drug Dev Technol 2025. [PMID: 39772938 DOI: 10.1089/adt.2024.120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
To optimize the formulation of docetaxel-zedoary oil magnetic solid lipid nanoparticles (DTX-ZTO-MSLN) using central composite design-response surface methodology. First, the formulation and preparation process of DTX-ZTO-MSLN were optimized via design-response surface methodology. The appearance, particle size, thermogravimetric, pH, iron content, magnetic strength, and in vitro drug release of DTX-ZTO-MSLN were subsequently examined. Finally, the antitumor effect of DTX-ZTO-MSLN on MCF-7 breast cancer cells was measured via the 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay. The optimized formulation was as follows: the mass ratio of soybean phospholipid to poloxamer 188 was 0.34, the mass ratio of DTX-ZTO to glycerol monostearate was 3.23, and 29.42 mL of water was used. The DTX-ZTO-MSLN prepared by the optimized method was clear and transparent, with good stability, with an iron content of 7.38%, and a saturation magnetization intensity of 7.05 A·m2·kg-1. The in vitro drug release was consistent with the Weibull model (R2 = 0.9992). Compared with zedoary turmeric oil and docetaxel, DTX-ZTO-MSLN had a much greater inhibitory effect on MCF-7 cells (p < 0.05). The optimized DTX-ZTO-MSLN meets the quality requirements for nanoemulsions. This study provides a theoretical basis for developing and applying DTX-ZTO-MSLN.
Collapse
Affiliation(s)
- Yujiao Hou
- Qiqihar Medical University, Qiqihar, China
- School of Medicine, Qilu Institute of Technology, Shandong, China
| | - Yuesheng Zhao
- Department of Mammography, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Jun Liu
- Qiqihar Medical University, Qiqihar, China
| | - Yanan Bao
- Qiqihar Medical University, Qiqihar, China
| | | | - Chunjie Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Wenjing Li
- Qiqihar Medical University, Qiqihar, China
| | - Bo Hong
- Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
4
|
Dhamija P, Mehata AK, Tamang R, Bonlawar J, Vaishali, Malik AK, Setia A, Kumar S, Challa RR, Koch B, Muthu MS. Redox-Sensitive Poly(lactic- co-glycolic acid) Nanoparticles of Palbociclib: Development, Ultrasound/Photoacoustic Imaging, and Smart Breast Cancer Therapy. Mol Pharm 2024; 21:2713-2726. [PMID: 38706253 DOI: 10.1021/acs.molpharmaceut.3c01086] [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] [Indexed: 05/07/2024]
Abstract
Breast cancer is one of the leading causes of mortality in women globally. The efficacy of breast cancer treatments, notably chemotherapy, is hampered by inadequate localized delivery of anticancer agents to the tumor site, resulting in compromised efficacy and increased systemic toxicity. In this study, we have developed redox-sensitive poly(lactic-co-glycolic acid) (PLGA) nanoparticles for the smart delivery of palbociclib (PLB) to breast cancer. The particle size of formulated PLB@PLGA-NPs (nonredox-sensitive) and RS-PLB@PLGA-NPs (redox-sensitive) NPs were 187.1 ± 1.8 nm and 193.7 ± 1.5 nm, respectively. The zeta potentials of nonredox-sensitive and redox-sensitive NPs were +24.99 ± 2.67 mV and +9.095 ± 1.87 mV, respectively. The developed NPs were characterized for morphological and various physicochemical parameters such as SEM, TEM, XRD, DSC, TGA, XPS, etc. The % entrapment efficiency of PLB@PLGA-NPs and RS-PLB@PLGA-NPs was found to be 85.48 ± 1.29% and 87.72 ± 1.55%, respectively. RS-PLB@PLGA-NPs displayed a rapid drug release at acidic pH and a higher GSH concentration compared to PLB@PLGA-NPs. The cytotoxicity assay in MCF-7 cells suggested that PLB@PLGA-NPs and RS-PLB@PLGA-NPs were 5.24-fold and 14.53-fold higher cytotoxic compared to the free PLB, respectively. Further, the cellular uptake study demonstrated that redox-sensitive NPs had significantly higher cellular uptake compared to nonredox-sensitive NPs and free Coumarin 6 dye. Additionally, AO/EtBr assay and reactive oxygen species analysis confirmed the superior activity of RS-PLB@PLGA-NPs over PLB@PLGA-NPs and free PLB. In vivo anticancer activity in dimethyl-benz(a)anthracene-induced breast cancer rats depicted that RS-PLB@PLGA-NPs was highly effective in reducing the tumor size, hypoxic tumor, and tumor vascularity compared to PLB@PLGA-NPs and free PLB. Further, hemocompatibility study reveals that the developed NPs were nonhemolytic to human blood. Moreover, an in vivo histopathology study confirmed that both nanoparticles were safe and nontoxic to the vital organs.
Collapse
Affiliation(s)
- Piyush Dhamija
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Rupen Tamang
- Genotoxicology and Cancer Biology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Jyoti Bonlawar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Vaishali
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Ankit Kumar Malik
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Aseem Setia
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Shailendra Kumar
- SATHI, Central Discovery Centre, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Ranadheer Reddy Challa
- Department of Pharmaceutical Science, School of Applied Sciences and Humanities, VIGNAN'S Foundation for Science, Technology and Research, Vadlamudi, Guntur, Andhra Pradesh 522213, India
| | - Biplob Koch
- Genotoxicology and Cancer Biology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
5
|
Sadwal S, Bharati S, Dar ZA, Kaur S. Chemopreventive potential of hydroethanolic Murraya koenigii leaves extract against DMBA induced breast carcinogenesis: In-silico and in-vivo study. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117124. [PMID: 37678421 DOI: 10.1016/j.jep.2023.117124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/28/2023] [Accepted: 09/03/2023] [Indexed: 09/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Murraya koenigii (MK), a member of the Rutaceae family and widely known as the curry-leaf tree, is indigenous to India, Sri Lanka, and other south Asian nations. It is a renowned medicinal herb because of the wide range of bioactive components found in its leaves, such as girinimbine, koenimbine, mahanimbine and mahanine among others. All these bioactive components make this plant beneficial for treating a variety of ailments and diseases. Biological and pharmacological activities of MK include anti-oxidant, anti-microbial, anti-ulcer, anti-helminthic, anti-malarial, anti-trichomonal, hepatoprotective, anti-diabetic, etc. AIM OF THE STUDY: The present study aimed to evaluate the possible protective effect of hydroethanolic Murraya koenigii leaves extract (HEMKLE) against 7,12-Dimethylbenz[a]anthracene (DMBA)-induced breast cancer in rats, which further paves the way for future breast cancer treatment. MATERIALS AND METHODS For the preparation of hydroethanolic Murraya koenigii leaves extract (HEMKLE), Murraya koenigii (MK) leaves were taken from the botanical garden of the Panjab University campus, Chandigarh, and authenticated from the Department of Botany, Panjab University (accession number 22417). The phytochemical characterization of HEMKLE was performed using liquid chromatography-mass spectrometry (LC-MS). Following this, an in-silico molecular docking analysis was performed using Maestro Schrodinger software, and an in-vivo study was conducted. For the in-vivo study, female SD rats were divided into four different groups. Group I (C), Group II (DMBA), Group III (HEMKLE), and Group IV (HEMKLE + DMBA). Histopathogy, oxidative and antioxidant status, immunohistochemistry of estrogen receptor-α, TUNEL assays, mRNA and protein expression of apoptotic pathway genes were conducted in in-vivo study. RESULTS In LC-MS, major phytochemical constituents including flavonoids and carbazole alkaloids were identified. In-silico docking study revealed the strong binding affinity between the identified compounds with caspase-3. Additionally, koenine displayed the highest binding affinity/minimum energy of -9.21 kcal/mol with 6BDV as compared to other phytochemicals. Furthermore, in-vivo experimentation revealed that HEMKLE administration in Group IV(HEMKLE + DMBA) significantly inhibits the tumor incidence and volume as compared to alone DMBA treated group. The antioxidant action of HEMKLE was proven from the in-vivo analysis of antioxidant marker enzymes, histopathology, immunohistochemistry of ER-α studies. Further, increase number of TUNEL positive cells was observed in co-treated animals as compared to alone DMBA treated animals. In Group IV (HEMKLE + DMBA), upregulated expression of pro-apoptotic genes and downregulated expression of anti-apoptotic gene were observed when compared to Group II(DMBA) suggested the apoptotic effect of HEMKLE. CONCLUSION The results of the present study provide clear evidence of the chemopreventive capabilities of HEMKLE in rats with DMBA-induced breast cancer. The observed outcomes could potentially be attributed to the existence of diverse phytochemicals within the HEMKLE.
Collapse
Affiliation(s)
- Shilpa Sadwal
- Department of Biophysics, Panjab University, Chandigarh, India.
| | - Sanjay Bharati
- Manipal College of Health Profession, Manipal Academy of Higher Education, Karnataka, India.
| | - Zahid Ahmad Dar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| | | |
Collapse
|
6
|
Alharbi DS, Albalawi SF, Alghrid ST, Alhwity BS, Qushawy M, Mortagi Y, El-Sherbiny M, Prabahar K, Elsherbiny N. Ginger Oil Nanoemulsion Formulation Augments Its Antiproliferative Effect in Ehrlich Solid Tumor Model. Foods 2023; 12:4139. [PMID: 38002196 PMCID: PMC10670723 DOI: 10.3390/foods12224139] [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: 10/18/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is a disease that is characterized by uncontrolled cell proliferation. Breast cancer is the most prevalent cancer among women. Ginger oil is a natural cancer fighter and anti-oxidant. However, the minimal absorption of ginger oil from the gastrointestinal tract accounts for its limited medicinal efficacy. The present study was designed to evaluate the efficacy of a nanoemulsion preparation of ginger oil on its oral bioavailability and in vivo anti-cancer efficacy. Ginger oil nanoemulsion was prepared by a high-pressure homogenization technique using different surfactants (Tween 20, 40, and 80). The prepared formulations were evaluated for droplet size, polydispersity index (PDI), zeta potential (ZP), pH, viscosity, and stability by calculating the creaming index percentage. The best formulation was evaluated for shape by TEM. The antitumor activity of the best nano-formulation was determined in comparison with the free oil using the in vivo Ehrlich solid tumor (EST) model. The prepared ginger oil nanoemulsion formulations exhibited acceptable droplet size in the range from 56.67 ± 3.10 nm to 357.17 ± 3.62 nm. A PDI of less than 0.5 indicates the homogeneity of size distribution. The oil globules possessed a negative charge ranging from -12.33 ± 1.01 to -39.33 ± 0.96 mV. The pH and viscosity were in the acceptable range. The TEM image of the best formulation appeared to be spherical with a small size. The ginger oil nanoemulsion reduced in vivo tumor volume and weight, extended animals' life span, and ameliorated liver and kidney function in EST-bearing mice. These effects were superior to using free ginger oil. Collectively, the present study demonstrated that the ginger oil nanoemulsion improved oral absorption with a subsequent enhancement of its anti-proliferative efficacy in vivo, suggesting a nano-formulation of ginger oil for better therapeutic outcomes in breast cancer patients.
Collapse
Affiliation(s)
- Danah S. Alharbi
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (D.S.A.); (S.F.A.); (S.T.A.); (B.S.A.)
| | - Shouq F. Albalawi
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (D.S.A.); (S.F.A.); (S.T.A.); (B.S.A.)
| | - Sarah T. Alghrid
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (D.S.A.); (S.F.A.); (S.T.A.); (B.S.A.)
| | - Basma S. Alhwity
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (D.S.A.); (S.F.A.); (S.T.A.); (B.S.A.)
| | - Mona Qushawy
- Department of Pharmaceutics, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, Sinai University, Alarish 45511, North Sinai, Egypt;
| | - Yasmin Mortagi
- Department of Pharmaceutics, Faculty of Pharmacy, Sinai University, Alarish 45511, North Sinai, Egypt;
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 13713, Saudi Arabia;
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Kousalya Prabahar
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Nehal Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
7
|
Albrakati A. Monosodium glutamate induces cortical oxidative, apoptotic, and inflammatory challenges in rats: the potential neuroprotective role of apigenin. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:24143-24153. [PMID: 36334201 DOI: 10.1007/s11356-022-23954-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/28/2022] [Indexed: 06/16/2023]
Abstract
Monosodium glutamate (MSG) is used as a flavor, and a taste enhancer was reported to evoke marked neuronal impairments. This study investigated the neuroprotective ability of flavonoid apigenin against neural damage in MSG-administered rats. Adult male rats were allocated into four groups: control, apigenin (20 mg/kg b.wt, orally), MSG (4 g/kg b.wt, orally), and apigenin + MSG at the aforementioned doses for 30 days. Regarding the levels of neurotransmitters, our results revealed that apigenin augmented the activity of acetylcholinesterase (AChE) markedly, and levels of brain monoamines (dopamine, norepinephrine, and serotonin) accompanied by lessening the activity of monoamine oxidase (MAO) as compared to MSG treatment. Moreover, apigenin counteracted the MSG-mediated oxidative stress by decreasing the malondialdehyde (MDA) levels together with elevating the glutathione (GSH) levels. In addition, pretreatment with apigenin induced notable increases in the activities of cortical superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and glutathione reductase (GR). Furthermore, apigenin attenuated the cortical inflammatory stress as indicated by lower levels of pro-inflammatory mediators such as interleukin-1 b (IL-1b), tumor necrosis factor-α (TNF-α), and nitric oxide (NO) as well as downregulated inducible nitric oxide synthase (iNOS) expression levels. Histopathological screening validated the abovementioned results and revealed that apigenin restored the distorted cytoarchitecture of the brain cortex. Thus, the present findings collectively suggest that apigenin exerted significant protection against MSG-induced neurotoxicity by enhancing the cellular antioxidant response and attenuating inflammatory machineries in the rat brain cortex.
Collapse
Affiliation(s)
- Ashraf Albrakati
- Department of Human Anatomy, College of Medicine, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
| |
Collapse
|
8
|
Agrawal MY, Gaikwad S, Srivastava S, Srivastava SK. Research Trend and Detailed Insights into the Molecular Mechanisms of Food Bioactive Compounds against Cancer: A Comprehensive Review with Special Emphasis on Probiotics. Cancers (Basel) 2022; 14:5482. [PMID: 36428575 PMCID: PMC9688469 DOI: 10.3390/cancers14225482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
In an attempt to find a potential cure for cancer, scientists have been probing the efficacy of the food we eat and its bioactive components. Over the decades, there has been an exponentially increasing trend of research correlating food and cancer. This review explains the molecular mechanisms by which bioactive food components exhibit anticancer effects in several cancer models. These bioactive compounds are mainly plant based or microbiome based. While plants remain the primary source of these phytochemicals, little is known about probiotics, i.e., microbiome sources, and their relationships with cancer. Thus, the molecular mechanisms underlying the anticancer effect of probiotics are discussed in this review. The principal mode of cell death for most food bioactives is found to be apoptosis. Principal oncogenic signaling axes such as Akt/PI3K, JAK/STAT, and NF-κB seem to be modulated due to these bioactives along with certain novel targets that provide a platform for further oncogenic research. It has been observed that probiotics have an immunomodulatory effect leading to their chemopreventive actions. Various foods exhibit better efficacy as complete extracts than their individual phytochemicals, indicating an orchestrated effect of the food components. Combining bioactive agents with available chemotherapies helps synergize the anticancer action of both to overcome drug resistance. Novel techniques to deliver bioactive agents enhance their therapeutic response. Such combinations and novel approaches are also discussed in this review. Notably, most of the food components that have been studied for cancer have shown their efficacy in vivo. This bolsters the claims of these studies and, thus, provides us with hope of discovering anticancer agents in the food that we eat.
Collapse
Affiliation(s)
- Manas Yogendra Agrawal
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
- Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Shreyas Gaikwad
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
- Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | | | - Sanjay K. Srivastava
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
- Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| |
Collapse
|
9
|
Alsagaby SA, Iqbal D, Ahmad I, Patel H, Mir SA, Madkhali YA, Oyouni AAA, Hawsawi YM, Alhumaydhi FA, Alshehri B, Alturaiki W, Alanazi B, Mir MA, Al Abdulmonem W. In silico investigations identified Butyl Xanalterate to competently target CK2α (CSNK2A1) for therapy of chronic lymphocytic leukemia. Sci Rep 2022; 12:17648. [PMID: 36271116 PMCID: PMC9587039 DOI: 10.1038/s41598-022-21546-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/28/2022] [Indexed: 01/18/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is an incurable malignancy of B-cells. In this study, bioinformatics analyses were conducted to identify possible pathogenic roles of CK2α, which is a protein encoded by CSNK2A1, in the progression and aggressiveness of CLL. Furthermore, various computational tools were used to search for a competent inhibitor of CK2α from fungal metabolites that could be proposed for CLL therapy. In CLL patients, high-expression of CSNK2A1 was associated with early need for therapy (n = 130, p < 0.0001) and short overall survival (OS; n = 107, p = 0.005). Consistently, bioinformatics analyses showed CSNK2A1 to associate with/play roles in CLL proliferation and survival-dependent pathways. Furthermore, PPI network analysis identified interaction partners of CK2α (PPI enrichment p value = 1 × 10-16) that associated with early need for therapy (n = 130, p < 0.003) and have been known to heavily impact on the progression of CLL. These findings constructed a rational for targeting CK2α for CLL therapy. Consequently, computational analyses reported 35 fungal metabolites out of 5820 (filtered from 19,967 metabolites) to have lower binding energy (ΔG: - 10.9 to - 11.7 kcal/mol) and better binding affinity (Kd: 9.77 × 107 M-1 to 3.77 × 108 M-1) compared with the native ligand (ΔG: - 10.8, Kd: 8.3 × 107 M--1). Furthermore, molecular dynamics simulation study established that Butyl Xanalterate-CK2α complex continuously remained stable throughout the simulation time (100 ns). Moreover, Butyl Xanalterate interacted with most of the catalytic residues, where complex was stabilized by more than 65% hydrogen bond interactions, and a significant hydrophobic interaction with residue Phe113. Here, high-expression of CSNK2A1 was implicated in the progression and poor prognosis of CLL, making it a potential therapeutic target in the disease. Butyl Xanalterate showed stable and strong interactions with CK2α, thus we propose it as a competitive inhibitor of CK2α for CLL therapy.
Collapse
Affiliation(s)
- Suliman A. Alsagaby
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Danish Iqbal
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Iqrar Ahmad
- grid.412233.50000 0001 0641 8393Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405 India
| | - Harun Patel
- grid.412233.50000 0001 0641 8393Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra 425405 India
| | - Shabir Ahmad Mir
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Yahya Awaji Madkhali
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Atif Abdulwahab A. Oyouni
- grid.440760.10000 0004 0419 5685Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia ,grid.440760.10000 0004 0419 5685Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Yousef M. Hawsawi
- grid.415310.20000 0001 2191 4301Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 40047, Jeddah, 21499 Kingdom of Saudi Arabia ,grid.411335.10000 0004 1758 7207College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh, 11533 Kingdom of Saudi Arabia
| | - Fahad A. Alhumaydhi
- grid.412602.30000 0000 9421 8094Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Bader Alshehri
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Wael Alturaiki
- grid.449051.d0000 0004 0441 5633Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, AL-Majmaah, 11952 Kingdom of Saudi Arabia
| | - Bader Alanazi
- grid.415277.20000 0004 0593 1832Biomedical Research Administration, Research Center, King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia ,Prince Mohammed bin Abdulaziz Medical City, AlJouf, Kingdom of Saudi Arabia
| | - Manzoor Ahmad Mir
- grid.412997.00000 0001 2294 5433Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Waleed Al Abdulmonem
- grid.412602.30000 0000 9421 8094Department of Pathology, College of Medicine, Qassim University, Qassim, Kingdom of Saudi Arabia
| |
Collapse
|