1
|
Zhou L, Li J, Ding C, Zhou Y, Xiao Z. Mechanistic Advances in Hypoglycemic Effects of Natural Polysaccharides: Multi-Target Regulation of Glycometabolism and Gut Microbiota Crosstalk. Molecules 2025; 30:1980. [PMID: 40363788 PMCID: PMC12073636 DOI: 10.3390/molecules30091980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Natural polysaccharides (NPs), as a class of bioactive macromolecules with multitarget synergistic regulatory potential, exhibit significant advantages in diabetes intervention. This review systematically summarizes the core hypoglycemic mechanisms of NPs, covering structure-activity relationships, integration of the gut microbiota-metabolism-immunity axis, and regulation of key signaling pathways. Studies demonstrate that the molecular weight, branch complexity, and chemical modifications of NPs mediate their hypoglycemic activity by influencing bioavailability and target specificity. NPs improve glucose metabolism through multiple pathways: activating insulin signaling, improving insulin resistance (IR), enhancing glycogen synthesis, inhibiting gluconeogenesis, and regulating gut microbiota homeostasis. Additionally, NPs protect pancreatic β-cell function via the nuclear factor E2-related factor 2 (Nrf2)/Antioxidant Response Element (ARE) antioxidant pathway and Toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) anti-inflammatory pathway. Clinical application of NPs still requires overcoming challenges such as resolving complex structure-activity relationships and dynamically integrating cross-organ signaling. Future research should focus on integrating multi-omics technologies (e.g., metagenomics, metabolomics) and organoid models to decipher the cross-organ synergistic action networks of NPs, and promote their translation from basic research to clinical applications.
Collapse
Affiliation(s)
- Liquan Zhou
- Homologous Innovation Laboratory of Medicine and Food, Hunan University of Chinese Medicine, Changsha 410208, China; (L.Z.); (J.L.); (C.D.)
- Hunan Engineering and Technology Research Center for Health Products and Life Science, Hunan University of Chinese Medicine, Changsha 410208, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jiani Li
- Homologous Innovation Laboratory of Medicine and Food, Hunan University of Chinese Medicine, Changsha 410208, China; (L.Z.); (J.L.); (C.D.)
- Hunan Engineering and Technology Research Center for Health Products and Life Science, Hunan University of Chinese Medicine, Changsha 410208, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chen Ding
- Homologous Innovation Laboratory of Medicine and Food, Hunan University of Chinese Medicine, Changsha 410208, China; (L.Z.); (J.L.); (C.D.)
- Hunan Engineering and Technology Research Center for Health Products and Life Science, Hunan University of Chinese Medicine, Changsha 410208, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yimiao Zhou
- Homologous Innovation Laboratory of Medicine and Food, Hunan University of Chinese Medicine, Changsha 410208, China; (L.Z.); (J.L.); (C.D.)
- Hunan Engineering and Technology Research Center for Health Products and Life Science, Hunan University of Chinese Medicine, Changsha 410208, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- Xiangyin Campus, Xiangxing College, Hunan University of Chinese Medicine, Yueyang 414615, China
| | - Zuowei Xiao
- Homologous Innovation Laboratory of Medicine and Food, Hunan University of Chinese Medicine, Changsha 410208, China; (L.Z.); (J.L.); (C.D.)
- Hunan Engineering and Technology Research Center for Health Products and Life Science, Hunan University of Chinese Medicine, Changsha 410208, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
2
|
Alves Nobre T, de Sousa AA, Pereira IC, Carvalho Pedrosa-Santos ÁM, Lopes LDO, Debia N, El-Nashar HAS, El-Shazly M, Islam MT, Castro E Sousa JMD, Torres-Leal FL. Bromelain as a natural anti-inflammatory drug: a systematic review. Nat Prod Res 2025; 39:1258-1271. [PMID: 38676413 DOI: 10.1080/14786419.2024.2342553] [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: 01/30/2024] [Revised: 03/09/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
Inflammation is a complex and necessary mechanism of an organ's response to biological, chemical and/or physical stimuli. In recent years, investigations on natural compounds with therapeutic actions for the treatment of different diseases have increased. Among these compounds, bromelain is highlighted, as a cysteine protease isolated from the Ananas comosus (pineapple) stem. This review aimed to evaluate the anti-inflammatory activity of bromelain, as well as its pathways on inflammatory mediators, through a systematic review with in vitro studies on different cell lines. The search was performed in PubMed, Science Direct, Scopus, Cochrane Library and Web of Science databases. Bromelain reduced IL-1β, IL-6 and TNF-α secretion when immune cells were already stimulated in an overproduction condition by proinflammatory cytokines, generating a modulation in the inflammatory response through prostaglandins reduction and activation of a cascade reactions that trigger neutrophils and macrophages, in addition to accelerating the healing process.
Collapse
Affiliation(s)
- Taline Alves Nobre
- Toxicological Genetics Research Laboratory (LAPGENIC), Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Athanara Alves de Sousa
- Toxicological Genetics Research Laboratory (LAPGENIC), Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Irislene Costa Pereira
- Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Álina Mara Carvalho Pedrosa-Santos
- Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Luana de Oliveira Lopes
- Toxicological Genetics Research Laboratory (LAPGENIC), Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Nicole Debia
- Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Heba A S El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- BioLuster Research Center, Dhaka, Bangladesh
| | - João Marcelo de Castro E Sousa
- Toxicological Genetics Research Laboratory (LAPGENIC), Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Francisco Leonardo Torres-Leal
- Metabolic Diseases, Exercise and Nutrition Research Group (DOMEN), Department of Biophysics and Physiology, Center for Health Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| |
Collapse
|
3
|
Karimi I, Ghowsi M, Mohammed LJ, Haidari Z, Nazari K, Schiöth HB. Inulin as a Biopolymer; Chemical Structure, Anticancer Effects, Nutraceutical Potential and Industrial Applications: A Comprehensive Review. Polymers (Basel) 2025; 17:412. [PMID: 39940613 PMCID: PMC11819723 DOI: 10.3390/polym17030412] [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: 01/01/2025] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Inulin is a versatile biopolymer that is non-digestible in the upper alimentary tract and acts as a bifidogenic prebiotic which selectively promotes gut health and modulates gut-organ axes through short-chain fatty acids and possibly yet-to-be-known interactions. Inulin usage as a fiber ingredient in food has been approved by the FDA since June 2018 and it is predicted that the universal inulin market demand will skyrocket in the near future because of its novel applications in health and diseases. This comprehensive review outlines the known applications of inulin in various disciplines ranging from medicine to industry, covering its benefits in gut health and diseases, metabolism, drug delivery, therapeutic pharmacology, nutrition, and the prebiotics industry. Furthermore, this review acknowledges the attention of researchers to knowledge gaps regarding the usages of inulin as a key modulator in the gut-organ axes.
Collapse
Affiliation(s)
- Isaac Karimi
- Research Group of Bioengineering and Biotechnology, Laboratory for Computational Physiology, Department of Biology, Faculty of Science, Razi University, P.O. Box 67149-67346, Kermanshah, Iran; (Z.H.); (K.N.)
- Department of Biology, Faculty of Science, Razi University, P.O. Box 67149-67346, Kermanshah, Iran;
| | - Mahnaz Ghowsi
- Department of Biology, Faculty of Science, Razi University, P.O. Box 67149-67346, Kermanshah, Iran;
| | - Layth Jasim Mohammed
- Department of Medical Microbiology, College of Medicine, Babylon University, Hilla City 51002, Babylon Governorate, Iraq;
| | - Zohreh Haidari
- Research Group of Bioengineering and Biotechnology, Laboratory for Computational Physiology, Department of Biology, Faculty of Science, Razi University, P.O. Box 67149-67346, Kermanshah, Iran; (Z.H.); (K.N.)
| | - Kosar Nazari
- Research Group of Bioengineering and Biotechnology, Laboratory for Computational Physiology, Department of Biology, Faculty of Science, Razi University, P.O. Box 67149-67346, Kermanshah, Iran; (Z.H.); (K.N.)
| | - Helgi B. Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, 751 24 Uppsala, Sweden
| |
Collapse
|
4
|
Manaer T, Sailike J, Sun X, Yeerjiang B, Nabi X. Therapeutic effects of composite probiotics derived from fermented camel milk on metabolic dysregulation and intestinal barrier integrity in type 2 diabetes rats. Front Pharmacol 2025; 15:1520158. [PMID: 39840100 PMCID: PMC11747018 DOI: 10.3389/fphar.2024.1520158] [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: 10/30/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025] Open
Abstract
Background In the Kazakh community of Xinjiang, China, fermented camel milk has been traditionally used to manage diabetes. This study evaluates the effects of composite probiotics derived from fermented camel milk (CPCM) on metabolic disturbances in a rat model of Type 2 diabetes (T2DM). Methods T2DM was induced in Wistar rats using streptozotocin. Experimental groups included a diabetic control, Metformin, and low- and high-dose CPCM. Measurements over 6 weeks included body weight (BW), fasting blood glucose (FBG), oral glucose tolerance test (OGTT), glycated hemoglobin (HbA1c), C-peptide (CP), lipid profiles, inflammatory markers, fecal short-chain fatty acids (SCFAs), and tight junction protein expression in colonic tissues. Results High-dose CPCM significantly increased BW by 22.2% (p < 0.05) and reduced FBG by 6.5 mmol/L (p < 0.001). The OGTT AUC decreased by 40.1% (p < 0.001), and HbA1c levels fell by 22.9% (p < 0.01). CP levels rose by 21.8% (p < 0.05). Lipid profiles improved: TC decreased by 40.0%, TG by 17.1%, and LDL-C by 30.4% (all p < 0.001). Fecal SCFAs, including acetate (75.4%, p < 0.001), methyl acetate (18.9%, p < 0.05), and butyrate (289.9%, p < 0.001), increased, with total SCFAs rising by 89.7% (p < 0.001). Inflammatory markers IL-1β (12.7%, p < 0.01), TNF-α (16.7%, p < 0.05), and IL-6 (17.3%, p < 0.01) were significantly reduced. Tight junction protein expression (ZO-1, occludin, claudin-1) and mucin (MUC2) in colonic tissues increased (p < 0.05). CPCM treatment also reduced serum total bile acids by 24.9%, while hepatic and fecal bile acids increased by 114.0% and 37.8% (all p < 0.001). CPCM lowered serum DAO, D-lactate, and LPS levels (all p < 0.001). mRNA levels of TGR5 and CYP7A1 in the liver, and TGR5 and FXR in the colon, were markedly elevated (all p < 0.001). Histological examinations revealed reduced pancreatic inflammation and hepatic steatosis, with restored colonic structure. Conclusion CPCM treatment significantly improved metabolic dysregulation in the T2DM rat model, reducing blood glucose and lipid levels, enhancing intestinal barrier function, and increasing insulin secretion. These findings highlight the therapeutic potential of CPCM in T2DM management and probiotics' role in metabolic health.
Collapse
Affiliation(s)
- Tabusi Manaer
- School of Pharmacy, Xinjiang Medical University, Urumchi, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumchi, China
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumchi, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumchi, China
| | | | - Xin Sun
- Srational for Drug Control and Medical Device Varification of Xinjiang Military Command, Urumchi, China
| | - Baheban Yeerjiang
- School of Pharmacy, Xinjiang Medical University, Urumchi, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumchi, China
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumchi, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumchi, China
| | - Xinhua Nabi
- School of Pharmacy, Xinjiang Medical University, Urumchi, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumchi, China
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumchi, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumchi, China
| |
Collapse
|
5
|
Rokonuzzman M, Bhuia MS, Al-Qaaneh AM, El-Nashar HAS, Islam T, Chowdhury R, Hasan Shanto H, Al Hasan MS, El-Shazly M, Torequl Islam M. Biomedical Perspectives of Citronellal: Biological Activities, Toxicological Profile and Molecular Mechanisms. Chem Biodivers 2025; 22:e202401973. [PMID: 39252577 DOI: 10.1002/cbdv.202401973] [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: 08/14/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/11/2024]
Abstract
Citronellal, known as rhodinal, is a naturally occurring monoterpenoid aldehyde distinctly found in the distilled oils of Cymbopogon species including C. marginatus, C. citratus, C. validus and C. winterianus family Gramineae. It is also obtained from eucalyptus, mentha, melissa, cinnamomum and allium. It is traditionally used in air freshener, cleaner, floor polishing, deodorants, moisturizing hand/body lotion, perfumes, and adhesives due to its lemon characteristic fragrance and therapeutic benefits. This study aimed to summarize the pharmacological activities and underlying mechanisms of citronellal against different diseases, as well as its toxicological profile. The data was collected from various reliable and authentic literatures by searching different academic search engines, including PubMed, Springer Link, Scopus, Wiley Online, Web of Science, ScienceDirect, and Google Scholar. The findings imply that citronellal demonstrated several pharmacological effects in various preclinical and pharmacological experimental systems. The results indicated that citronellal demonstrated antioxidant, anti-inflammatory, antibacterial, antifungal, anthelminthic, and anticancer effects with beneficial effects in neurological and cardiovascular diseases. Our findings also indicated the toxic level of the phytochemical. In conclusion, it has been proposed that citronellal has the capability to serve as a hopeful therapeutic agent, so further extensive clinical research is necessary to develop it as a reliable drug.
Collapse
Affiliation(s)
- Md Rokonuzzman
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj Dhaka, 8100, Bangladesh
| | - Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj Dhaka, 8100, Bangladesh
| | - Ayman M Al-Qaaneh
- Department of Allied Health Sciences, Al-Balqa Applied University (BAU), Al-Salt, 19117, Jordan
| | - Heba A S El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt E-mai
| | - Tawhida Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Hasibul Hasan Shanto
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt E-mai
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj Dhaka, 8100, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, 9208, Bangladesh
| |
Collapse
|
6
|
Lou Y, Wen X, Song S, Zeng Y, Huang L, Xie Z, Shao T, Wen C. Dietary pectin and inulin: A promising adjuvant supplement for collagen-induced arthritis through gut microbiome restoration and CD4 + T cell reconstitution. J Nutr Biochem 2024; 133:109699. [PMID: 38972609 DOI: 10.1016/j.jnutbio.2024.109699] [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: 10/09/2023] [Revised: 05/17/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
Dietary strategies rich in fiber have been demonstrated to offer benefits to individuals afflicted with rheumatoid arthritis (RA). However, the specific mechanisms through which a high-fiber diet (HFD) mitigates RA's autoimmunity remain elusive. Herein, we investigate the influence of pectin- and inulin-rich HFD on collagen-induced arthritis (CIA). We establish that HFD significantly alleviates arthritis in CIA mice by regulating the Th17/Treg balance. The rectification of aberrant T cell differentiation by the HFD is linked to the modulation of gut microbiota, augmenting the abundance of butyrate in feces. Concurrently, adding butyrate to the drinking water mirrors the HFD's impact on ameliorating CIA, encompassing arthritis mitigation, regulating intestinal barrier integrity, and restoring the Th17/Treg equilibrium. Butyrate reshapes the metabolic profile of CD4+ T cells in an AMPK-dependent manner. Our research underscores the importance of dietary interventions in rectifying gut microbiota for RA management and offers an explanation of how diet-derived microbial metabolites influence RA's immune-inflammatory-reaction.
Collapse
Affiliation(s)
- Yu Lou
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianghui Wen
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China; Department of Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Siyue Song
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yufeng Zeng
- Department of Clinical Medicine, The 2ND Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Huang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijun Xie
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tiejuan Shao
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Chengping Wen
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
7
|
Bhirud D, Bhattacharya S, Prajapati BG. Bioengineered carbohydrate polymers for colon-specific drug release: Current trends and future prospects. J Biomed Mater Res A 2024; 112:1860-1872. [PMID: 38721841 DOI: 10.1002/jbm.a.37732] [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: 03/10/2024] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 09/03/2024]
Abstract
The worldwide health burden of colorectal cancer is still substantial, and traditional chemotherapeutic drugs sometimes have poor selectivity, which can result in systemic toxicity and unfavorable side effects. For colon-specific medication delivery, bioengineered carbohydrate polymers have shown promise as carriers. They may enhance treatment effectiveness while minimizing systemic exposure and associated side effects. The unique properties of these manufactured or naturally occurring biopolymers, such as hyaluronic acid, chitosan, alginate, and pectin, enable targeted medicine release. These qualities can be changed to meet the physiological needs of the colon. In the context of colorectal cancer therapy, this article provides a comprehensive overview of current developments and prospective future directions in the field of bioengineered carbohydrate polymer synthesis for colon-specific drug delivery. We discuss numerous techniques for achieving colon-targeted drug release, including enzyme-sensitive polymers, pH-responsive devices, and microbiota-activated processes. To increase tumor selectivity and cellular uptake, we also examine the inclusion of active targeting approaches, such as conjugating specific ligands. Furthermore, we discuss the potential of combination treatment strategies, which use the coadministration of numerous therapeutic medications to target multiple pathways implicated in cancer growth and address drug resistance mechanisms. We address recent biomimetic advances that potentially improve the biocompatibility, cellular uptake, and tumor penetration of carbohydrate polymer-based nanocarriers. These methods involve protein corona engineering and cell membrane coating. Furthermore, we look at the possibility of intelligent and sensitive systems that may adjust their behaviors in response to certain inputs or feedback loops, allowing for precise and regulated drug distribution.
Collapse
Affiliation(s)
- Darshan Bhirud
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM's NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM's NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Shree S.K. Patel College of Pharmaceutical Education & Research, Mehsana, Gujarat, India
| |
Collapse
|
8
|
Hassan SH, El-Nashar HAS, Rahman MA, Polash JI, Bappi MH, Mondal M, Abdel-Maksoud MA, Malik A, Aufy M, El-Shazly M, Islam MT. Sclareol antagonizes the sedative effect of diazepam in thiopental sodium-induced sleeping animals: In vivo and in silico studies. Biomed Pharmacother 2024; 176:116939. [PMID: 38870629 DOI: 10.1016/j.biopha.2024.116939] [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: 03/27/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Sclareol (SCL), a labdane diterpene compound found in Salvia sclarea L., exhibited therapeutic effects. This study investigated the potential interaction between SCL and diazepam (DZP) in modulating sedation in the thiopental sodium-induced sleeping animal model, supported by in-silico molecular docking analysis. METHODS The control, sclareol (5, 10 and 20 mg/kg), and the reference drugs [diazepam: 3 mg/kg and Caffeine (CAF): 10 mg/kg] were used in male albino mice. Then, sodium thiopental (40 mg/kg, i.p.) was administrated to induce sleep. The latent period, percentage of sleep incidence and modulation of latency were measured. Further, homology modeling of human γ-aminobutyric acid (GABA) was conducted examine the binding mode of GABA interaction with SCL, DZP, and CAF compounds RESULTS: SCL (low dose) slightly increased the sleep latency, while the higher dose significantly prolonged sleep latency. DZP, a GABAA receptor agonist, exhibited strong sleep-inducing properties, reducing sleep latency, and increasing sleeping time. Caffeine (CAF) administration prolonged sleep latency and reduced sleeping time, consistent with its stimulant effects. The combination treatments involving SCL, DZP, and CAF showed mixed effects on sleep parameters. The molecular docking revealed good binding affinities of SCL, DZP, and CAF for GABAA receptor subunits A2 and A5. CONCLUSIONS Our findings highlighted the complex interplay between SCL, DZP, and CAF in regulating sleep behaviors and provided insights into potential combination therapies for sleep disorders.
Collapse
Affiliation(s)
- Sm Hafiz Hassan
- Department of Chemistry and Biochemistry, Miami University, USA
| | - Heba A S El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt.
| | - Md Anisur Rahman
- Department of Pharmacy, Islamic University, Kushtia 7003, Bangladesh
| | | | - Mehedi Hasan Bappi
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Milon Mondal
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | | | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Saudi Arabia
| | - Mohammed Aufy
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Austria.
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; Pharmacy Discipline, Khulna University, Khulna 9208, Bangladesh; BioLuster Research Center, Gopalganj, Dhaka 8100, Bangladesh
| |
Collapse
|
9
|
Juhi UH, El-Nashar HAS, Al Faruq A, Bhuia MS, Sultana I, Alam S, Abuyousef F, Saleh N, El-Shazly M, Islam MT. Phytochemical analysis and biological investigation of Cheilanthes tenuifolia (Burm.f.) Swartz. Front Pharmacol 2024; 15:1366889. [PMID: 38638865 PMCID: PMC11024464 DOI: 10.3389/fphar.2024.1366889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/07/2024] [Indexed: 04/20/2024] Open
Abstract
Introduction: Cheilanthes tenuifolia is an evergreen ornamental small fern, belonging to the family Pteridaceae, that grows in warm and rocky regions worldwide. Many species of Cheilanthes genus are evidently endowed with important phytochemicals and bioactivities. This study aimed to perform a preliminary phytochemical analysis of Cheilanthes tenuifolia leaves alongside an evaluation of free radical scavenging, anti-inflammatory, antimicrobial, and clot lysis activities of extract fractions. Materials and methods: A preliminary phytochemical analysis was done after fractionation of ethanolic extract (ECT) with n-hexane (HCT) and chloroform (CCT). Then, 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging, egg albumin and RBC membrane stabilization tests, disc diffusion, and human blood clot lysis assays were performed. Results: Phytochemical investigations suggested that the plant is rich in alkaloids, glycosides, tannins, and flavonoids. All obtained fractions exhibited concentration-dependent radical scavenging, inhibition of egg protein denaturation and RBC membrane lysis capacities. Except for antifungal tests, ECT exhibited better DPPH radical scavenging, anti-inflammatory, antibacterial, and clot lysis capacities than HCT and CCT fractions. However, all fractions exhibited a mild anti-inflammatory activity. Conclusion: C. tenuifolia might be a good source of antioxidant, anti-microbial, and anti-atherothrombotic agents. Further studies are required to isolate and characterize the active principles liable for each bioactivity, along with possible molecular interactions.
Collapse
Affiliation(s)
- Umme Habiba Juhi
- Department of Pharmacy, Southern University Bangladesh, Chattogram, Bangladesh
| | - Heba A. S. El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Abdullah Al Faruq
- Department of Pharmacy, Southern University Bangladesh, Chattogram, Bangladesh
| | - Md. Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Bioluster Research Center, Dhaka, Bangladesh
| | - Irin Sultana
- Department of Pharmacy, Southern University Bangladesh, Chattogram, Bangladesh
| | - Syedul Alam
- Forest Botany Division, Bangladesh Forest Research Institute (BFRI), Chattogram, Bangladesh
| | - Farah Abuyousef
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Na’il Saleh
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Bioluster Research Center, Dhaka, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, Bangladesh
| |
Collapse
|