1
|
Li R, Wang Y, Lao Y, You C, Qing L, Guan X, Wang J, Li X, Li Q, Liu S, Dong Z. Effect and Mechanism of Aloin in Ameliorating Chronic Prostatitis/Chronic Pelvic Pain Syndrome: Network Pharmacology and Experimental Verification. Drug Des Devel Ther 2025; 19:1945-1969. [PMID: 40110504 PMCID: PMC11920635 DOI: 10.2147/dddt.s473678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025] Open
Abstract
Purpose This research aims to investigate the role and potential mechanisms of Aloin in Chronic Prostatitis/Chronic Pelvic Pain Syndrome (CP/CPPS) through network pharmacology and experimental approaches. Methods Using network pharmacology methods, potential targets of Aloin and targets related to CP/CPPS were screened from public databases. The protein-protein interaction (PPI) network, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed to predict the core targets and pathways of Aloin against CP/CPPS. The effects of Aloin in ameliorating CP/CPPS were verified in animal experiments. Results A total of 235 genes interacting with Aloin in CP/CPPS were identified. PPI network analysis revealed five core targets: AKT1, EGFR, ESR1, HSP90AA1, and SRC. GO analysis yielded 2916 enrichment results, with 2562 related to Biological Process (BP), 94 to Cellular Component (CC), and 260 to Molecular Function (MF). KEGG pathway analysis identified 172 pathways. Molecular docking confirmed stable binding between Aloin and core targets. Molecular dynamics simulations further validated binding stability by analyzing Root Mean Square Deviation (RMSD), Root Mean Square Fluctuation (RMSF), Radius of Gyration (Rg), hydrogen bonds, Solvent Accessible Surface Area (SASA), and Gibbs free energy of Aloin-target complexes. Experimental validation showed that Aloin alleviated pain, reduced inflammatory factors, and decreased oxidative stress in a rat model of CP/CPPS. The qRT-PCR results showed that Aloin intervention reduced the mRNA expression of AKT1, EGFR, HSP90AA1, and SRC, while increasing ESR1 mRNA expression. These changes may underlie its therapeutic effects in CP/CPPS. Conclusion Our study revealed that Aloin exerts a beneficial effect on mitigating the pain symptoms associated with CP/CPPS, ameliorating inflammation, and reducing oxidative stress. Through network pharmacology, potential targets and signaling pathways were identified, suggesting the therapeutic promise of Aloin for CP/CPPS. These findings advocate for further exploration into its clinical efficacy and mechanistic underpinnings in the treatment of CP/CPPS.
Collapse
Affiliation(s)
- Rongxin Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, 730000, People's Republic of China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, Gansu, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, People's Republic of China
| | - Yanan Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, 730000, People's Republic of China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, Gansu, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, People's Republic of China
| | - Yongfeng Lao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, 730000, People's Republic of China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, Gansu, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, People's Republic of China
| | - Chengyu You
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, 730000, People's Republic of China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, Gansu, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, People's Republic of China
| | - Liangliang Qing
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, 730000, People's Republic of China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, Gansu, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, People's Republic of China
| | - Xin Guan
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, 730000, People's Republic of China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, Gansu, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, People's Republic of China
| | - Jian Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, 730000, People's Republic of China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, Gansu, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, People's Republic of China
| | - Xiaolong Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, 730000, People's Republic of China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, Gansu, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, People's Republic of China
| | - Qingchao Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, 730000, People's Republic of China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, Gansu, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, People's Republic of China
| | - Shuai Liu
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, 730000, People's Republic of China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, Gansu, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, People's Republic of China
| | - Zhilong Dong
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, 730000, People's Republic of China
- Gansu Province Key Laboratory of Urological Diseases, Lanzhou, Gansu, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, Gansu, 730000, People's Republic of China
| |
Collapse
|
2
|
Zheng Y, Gao Y, Zhu W, Bai XG, Qi J. Advances in molecular agents targeting toll-like receptor 4 signaling pathways for potential treatment of sepsis. Eur J Med Chem 2024; 268:116300. [PMID: 38452729 DOI: 10.1016/j.ejmech.2024.116300] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/23/2024] [Accepted: 03/01/2024] [Indexed: 03/09/2024]
Abstract
Sepsis is a systemic inflammatory response syndrome caused by an infection. Toll-like receptor 4 (TLR4) is activated by endogenous molecules released by injured or necrotic tissues. Additionally, TLR4 is remarkably sensitive to infection of various bacteria and can rapidly stimulate host defense responses. The TLR4 signaling pathway plays an important role in sepsis by activating the inflammatory response. Accordingly, as part of efforts to improve the inflammatory response and survival rate of patients with sepsis, several drugs have been developed to regulate the inflammatory signaling pathways mediated by TLR4. Inhibition of TLR4 signal transduction can be directed toward either TLR4 directly or other proteins in the TLR4 signaling pathway. Here, we review the advances in the development of small-molecule agents and peptides targeting regulation of the TLR4 signaling pathway, which are characterized according to their structural characteristics as polyphenols, terpenoids, steroids, antibiotics, anthraquinones, inorganic compounds, and others. Therefore, regulating the expression of the TLR4 signaling pathway and modulating its effects has broad prospects as a target for the treatment of lung, liver, kidneys, and other important organs injury in sepsis.
Collapse
Affiliation(s)
- Yunyun Zheng
- Medicine College of Pingdingshan University, Pingdingshan, Henan, 467000, China
| | - Yingying Gao
- Medicine College of Pingdingshan University, Pingdingshan, Henan, 467000, China
| | - Weiru Zhu
- Medicine College of Pingdingshan University, Pingdingshan, Henan, 467000, China
| | - Xian-Guang Bai
- Medicine College of Pingdingshan University, Pingdingshan, Henan, 467000, China.
| | - Jinxu Qi
- Medicine College of Pingdingshan University, Pingdingshan, Henan, 467000, China.
| |
Collapse
|
3
|
Araújo GDS, Brilhante RSN, Rocha MGD, Aguiar LD, Castelo-Branco DDSCM, Guedes GMDM, Sidrim JJC, Pereira Neto WA, Rocha MFG. Anthraquinones against Cryptococcus neoformans sensu stricto: antifungal interaction, biofilm inhibition and pathogenicity in the Caenorhabditis elegans model. J Med Microbiol 2024; 73. [PMID: 38530134 DOI: 10.1099/jmm.0.001815] [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: 03/27/2024] Open
Abstract
Introduction. Cryptococcal biofilms have been associated with persistent infections and antifungal resistance. Therefore, strategies, such as the association of natural compounds and antifungal drugs, have been applied for the prevention of biofilm growth. Moreover, the Caenorhabditis elegans pathogenicity model has been used to investigate the capacity to inhibit the pathogenicity of Cryptococcus neoformans sensu stricto.Hypothesis. Anthraquinones and antifungals are associated with preventing C. neoformans sensu stricto biofilm formation and disrupting these communities. Antraquinones reduced the C. neoformans sensu stricto pathogenicity in the C. elegans model.Aim. This study aimed to evaluate the in vitro interaction between aloe emodin, barbaloin or chrysophanol and itraconazole or amphotericin B against growing and mature biofilms of C. neoformans sensu stricto.Methodology. Compounds and antifungal drugs were added during biofilm formation or after 72 h of growth. Then, the metabolic activity was evaluated by the MTT reduction assay, the biomass by crystal-violet staining and the biofilm morphology by confocal laser scanning microscopy. C. neoformans sensu stricto's pathogenicity was investigated using the nematode C. elegans. Finally, pathogenicity inhibition by aloe emodin, barbarloin and chrysophanol was investigated using this model.Results. Anthraquinone-antifungal combinations affected the development of biofilms with a reduction of over 60 % in metabolic activity and above 50 % in biomass. Aloe emodin and barbaloin increased the anti-biofilm activity of antifungal drugs. Chrysophanol potentiated the effect of itraconazole against C. neoformans sensu stricto biofilms. The C. elegans mortality rate reached 76.7 % after the worms were exposed to C. neoformans sensu stricto for 96 h. Aloe emodin, barbaloin and chrysophanol reduced the C. elegans pathogenicity with mortality rates of 61.12 %, 65 % and 53.34 %, respectively, after the worms were exposed for 96 h to C. neoformans sensu stricto and these compounds at same time.Conclusion. These results highlight the potential activity of anthraquinones to increase the effectiveness of antifungal drugs against cryptococcal biofilms.
Collapse
Affiliation(s)
- Géssica Dos Santos Araújo
- Postgraduate Program in Veterinary Sciences, School of Veterinary, State University of Ceará. Dr. Silas Munguba, 1700, Campus do Itaperi, CEP: 60714-903, Fortaleza, Ceará, Brazil
| | - Raimunda Sâmia Nogueira Brilhante
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Coronel Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Maria Gleiciane da Rocha
- Postgraduate Program in Veterinary Sciences, School of Veterinary, State University of Ceará. Dr. Silas Munguba, 1700, Campus do Itaperi, CEP: 60714-903, Fortaleza, Ceará, Brazil
| | - Lara de Aguiar
- Postgraduate Program in Veterinary Sciences, School of Veterinary, State University of Ceará. Dr. Silas Munguba, 1700, Campus do Itaperi, CEP: 60714-903, Fortaleza, Ceará, Brazil
| | - Débora de Souza Collares Maia Castelo-Branco
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Coronel Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Glaucia Morgana de Melo Guedes
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Coronel Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - José Júlio Costa Sidrim
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Coronel Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Waldemiro Aquino Pereira Neto
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Coronel Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Marcos Fábio Gadelha Rocha
- Postgraduate Program in Veterinary Sciences, School of Veterinary, State University of Ceará. Dr. Silas Munguba, 1700, Campus do Itaperi, CEP: 60714-903, Fortaleza, Ceará, Brazil
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará. Rua Coronel Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| |
Collapse
|
4
|
Peng J, Li S, Han M, Gao F, Qiao L, Tian Y. SNHG1/miR-21 axis mediates the cardioprotective role of aloin in sepsis through modulating cardiac cell viability and inflammatory responses. J Clin Lab Anal 2023; 37:e24985. [PMID: 37950500 PMCID: PMC10749494 DOI: 10.1002/jcla.24985] [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: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Aloin has cardioprotective effects, however, its cardioprotective role in sepsis remains unclear. This study aimed to analyze whether aloin could prevent sepsis-related myocardial damage and explore the underlying mechanisms by examining the expression of long-noncoding RNA (lncRNA) SNHG1 and microRNA-21 (miR-21). METHODS The interaction of SNHG1 with miR-21 was identified by dual-luciferase reporter assay. The levels of SNHG1 and miR-21 were measured by real-time quantitative PCR. The cardioprotective function of aloin was assessed in a sepsis animal model, which was induced by cecal ligation and puncture, and in a myocardial injury cell model in H9C2 cells stimulated by lipopolysaccharide. Myocardial injury biomarker levels and hemodynamic indicators in mice model were measured to evaluate cardiac function. The viability of H9C2 cells was assessed by cell counting kit-8 assay. Inflammatory cytokine levels were examined by an ELISA method. RESULTS Decreased SNHG1 and increased miR-21 were found in sepsis patients with cardiac dysfunction, and they were negatively correlated. Aloin significantly attenuated myocardial damage and inflammatory responses of mice model, and increased the viability and suppressed inflammation in H9C2 cell model. In addition, SNHG1 expression was upregulated and miR-21 expression was downregulated by aloin in both mice and cell models. Moreover, in mice and cell models, SNHG1/miR-21 axis affected sepsis-related myocardial damage, and mediated the cardioprotective effects of aloin. CONCLUSION Our findings indicated that aloin exerts protective effects in sepsis-related myocardial damage through regulating cardiac cell viability and inflammatory responses via regulating the SNHG1/miR-21 axis.
Collapse
Affiliation(s)
- Jin Peng
- Intensive Care UnitShengli Oilfield Central HospitalDongyingShandongChina
| | - Shuyuan Li
- Intensive Care UnitShengli Oilfield Central HospitalDongyingShandongChina
| | - Maozhi Han
- Department of PharmacyThe 80th Army HospitalWeifangShandongChina
| | - Feng Gao
- Applied Pharmacology LaboratoryWeifang Medical CollegeWeifangShandongChina
| | - Lujun Qiao
- Intensive Care UnitShengli Oilfield Central HospitalDongyingShandongChina
| | - Yonggang Tian
- Intensive Care UnitShengli Oilfield Central HospitalDongyingShandongChina
| |
Collapse
|
5
|
Qun T, Zhou T, Hao J, Wang C, Zhang K, Xu J, Wang X, Zhou W. Antibacterial activities of anthraquinones: structure-activity relationships and action mechanisms. RSC Med Chem 2023; 14:1446-1471. [PMID: 37593578 PMCID: PMC10429894 DOI: 10.1039/d3md00116d] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/24/2023] [Indexed: 08/19/2023] Open
Abstract
With the increasing prevalence of untreatable infections caused by antibiotic-resistant bacteria, the discovery of new drugs from natural products has become a hot research topic. The antibacterial activity of anthraquinones widely distributed in traditional Chinese medicine has attracted much attention. Herein, the structure and activity relationships (SARs) of anthraquinones as bacteriostatic agents are reviewed and elucidated. The substituents of anthraquinone and its derivatives are closely related to their antibacterial activities. The stronger the polarity of anthraquinone substituents is, the more potent the antibacterial effects appear. The presence of hydroxyl groups is not necessary for the antibacterial activity of hydroxyanthraquinone derivatives. Substitution of di-isopentenyl groups can improve the antibacterial activity of anthraquinone derivatives. The rigid plane structure of anthraquinone lowers its water solubility and results in the reduced activity. Meanwhile, the antibacterial mechanisms of anthraquinone and its analogs are explored, mainly including biofilm formation inhibition, destruction of the cell wall, endotoxin inhibition, inhibition of nucleic acid and protein synthesis, and blockage of energy metabolism and other substances.
Collapse
Affiliation(s)
- Tang Qun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
| | - Tiantian Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University 440113 Guangzhou China
| | - Jiongkai Hao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
| | - Chunmei Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Keyu Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Jing Xu
- Huanghua Agricultural and Rural Development Bureau Bohai New Area 061100 Hebei China
| | - Xiaoyang Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Wen Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| |
Collapse
|
6
|
Su J, Zhou F, Wu S, Tong Z. Research Progress on Natural Small-Molecule Compounds for the Prevention and Treatment of Sepsis. Int J Mol Sci 2023; 24:12732. [PMID: 37628912 PMCID: PMC10454676 DOI: 10.3390/ijms241612732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Sepsis is a serious disease with high mortality and has been a hot research topic in medical research in recent years. With the continuous reporting of in-depth research on the pathological mechanisms of sepsis, various compounds have been developed to prevent and treat sepsis. Natural small-molecule compounds play vital roles in the prevention and treatment of sepsis; for example, compounds such as resveratrol, emodin, salidroside, ginsenoside, and others can modulate signaling through the NF-κB, STAT3, STAT1, PI3K, and other pathways to relieve the inflammatory response, immunosuppression, and organ failure caused by sepsis. Here, we discuss the functions and mechanisms of natural small-molecule compounds in preventing and treating sepsis. This review will lay the theoretical foundation for discovering new natural small-molecule compounds that can potentially prevent and treat sepsis.
Collapse
|
7
|
Kim C, Jeong YH, Kim N, Ryu SH, Bae JS. Hepatoprotective functions of jujuboside B. J Nat Med 2023; 77:87-95. [PMID: 36064835 DOI: 10.1007/s11418-022-01648-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/14/2022] [Indexed: 01/06/2023]
Abstract
Jujuboside B (JB) found in the seeds of Zizyphi Spinosi Semen possesses pharmacological functions, such as anti-inflammatory, antiplatelet aggregation, and antianxiety potentials. This study evaluated the effect of JB on liver failure in cecal ligation and puncture (CLP)-induced sepsis. First, we observed histopathological changes in the liver by optical microscopy and the activity of enzymes in serum such as alanine aminotransferase (ALT) and aspartate aminotransferase (AST). We further measured the levels of interleukin (IL)-1β, tumor necrosis factor (TNF)-α, nitric oxide (NO), and antioxidative parameters in liver homogenate. The expression of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2), and glucocorticoid receptor (GR) in the liver was observed by Western blotting. CLP enhanced the migration of inflammatory cells, ALT and AST concentrations, and necrosis, which were reduced by JB. In addition, JB reduced 11β-HSD2 expression and levels of inflammatory mediators (TNF-α, IL-1β, and NO) in the liver, increased GR expression, enhanced endogenous antioxidative capacity. These results further suggest that JB may protect the liver against CLP-induced damage by regulating anti-inflammatory responses, downregulating 11β-HSD2 expression and antioxidation, and up-regulating GR expression.
Collapse
Affiliation(s)
- Chaeyeong Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 41566, Republic of Korea
| | - Yun Hee Jeong
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine, Daegu, 41062, Republic of Korea
| | - Nayeon Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 41566, Republic of Korea
| | - Soo Ho Ryu
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 41566, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 41566, Republic of Korea.
| |
Collapse
|
8
|
Mahomoodally MF, Aumeeruddy MZ, Legoabe LJ, Dall’Acqua S, Zengin G. Plants' bioactive secondary metabolites in the management of sepsis: Recent findings on their mechanism of action. Front Pharmacol 2022; 13:1046523. [PMID: 36588685 PMCID: PMC9800845 DOI: 10.3389/fphar.2022.1046523] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Sepsis is a severe inflammatory response to systemic infection and is a threatening cause of death in intensive care units. In recent years, a number of studies have been conducted on the protective effect of natural products against sepsis-induced organ injury. However, a comprehensive review of these studies indicating the mechanisms of action of the bioactive compounds is still lacking. In this context, this review aimed to provide an updated analysis of the mechanism of action of plants' secondary metabolites in the management of sepsis. Scopus, Science Direct, Google Scholar, and PubMed were searched from inception to July 2022. A variety of secondary metabolites were found to be effective in sepsis management including allicin, aloin, cepharanthine, chrysin, curcumin, cyanidin, gallic acid, gingerol, ginsenoside, glycyrrhizin, hesperidin, kaempferol, narciclasine, naringenin, naringin, piperine, quercetin, resveratrol, rosmarinic acid, shogaol, silymarin, sulforaphane, thymoquinone, umbelliferone, and zingerone. The protective effects exerted by these compounds can be ascribed to their antioxidant properties as well as induction of endogenous antioxidant mechanisms, and also via the downregulation of inflammatory response and reduction of biochemical and inflammatory markers of sepsis. These findings suggest that these secondary metabolites could be of potential therapeutic value in the management of sepsis, but human studies must be performed to provide strength to their potential clinical relevance in sepsis-related morbidity and mortality reduction.
Collapse
Affiliation(s)
- Mohamad Fawzi Mahomoodally
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam,Faculty of Natural Sciences, Duy Tan University, Da Nang, Vietnam,*Correspondence: Mohamad Fawzi Mahomoodally, ; Stefano Dall’Acqua,
| | | | - Lesetja Jan Legoabe
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen), North West University, Potchefstroom, South Africa
| | - Stefano Dall’Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy,*Correspondence: Mohamad Fawzi Mahomoodally, ; Stefano Dall’Acqua,
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University, Campus, Konya, Turkey
| |
Collapse
|
9
|
Lee IC, Bae JS. Inhibitory effects of aloin on lipopolysaccharide-induced severe inflammatory responses. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2022; 24:987-999. [PMID: 35023793 DOI: 10.1080/10286020.2022.2026932] [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: 02/15/2020] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 06/14/2023]
Abstract
Aloin is the main anthraquinone glycoside from Aloe species. Here, the anti-inflammatory functions of aloin against lipopolysaccharide (LPS)-induced vascular inflammatory responses were tested in endothelial cells or mice such as permeability, expressions of cell adhesion molecule (CAM), migration of leukocytes and lethality. Aloin was found to inhibit LPS-induced barrier disruption, CAM expression, and neutrophil adhesion/transendothelial migration to endothelial cells. Furthermore, aloin inhibited LPS-induced hyperpermeability, leukocyte migration, lethality in vivo. These results suggest that aloin has anti-inflammatory activities against LPS, thereby supporting its usefulness as a treatment for vascular inflammatory.
Collapse
Affiliation(s)
- In-Chul Lee
- Department of Cosmetic Science and Technology, Seowon University, Cheongju 28674, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
10
|
Kim GO, Kim N, Song GY, Bae JS. Inhibitory Activities of Rare Ginsenoside Rg4 on Cecal Ligation and Puncture-Induced Sepsis. Int J Mol Sci 2022; 23:ijms231810836. [PMID: 36142743 PMCID: PMC9505814 DOI: 10.3390/ijms231810836] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Sepsis is an uncontrolled response to inflammatory infection and is associated with high levels of mortality and morbidity. Rg4 is a rare ginsenoside mainly found in the leaves of Panax ginseng C. A. Meyer and the major protopanaxatriol-type ginsenoside of black ginseng. In this study, we determined whether Rg4 affects cecal ligation and puncture (CLP)-induced sepsis. Animals were separated into the following six groups: control group, CLP-operated group, CLP plus maslinic acid (MA), and CLP plus Rg4 (5, 10, or 15 mg/kg). Survival rate, body weight changes, inflammatory cytokines, and histological analyses were assessed. Human endothelial cells were activated with the high-mobility group box 1 (HMGB1) protein and Rg4. Cell viability was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Enzyme-linked immunosorbent assay (ELISA) and Western blot analysis were used to assess inflammation and gene expression, respectively. After CLP surgery, the Rg4-administered group exhibited a higher survival rate and body weight compared with the untreated control group. Rg4 treatment reduced cytokine levels, including tumor necrosis factor (TNF)-α and interleukin (IL)-1β, as well as nitric oxide (NO) levels and renal inflammation. After Rg4 treatment of HMGB1-activated cells, the expressions of toll-like receptor (TLR) 4 and TNF-α were decreased, and the activation of phosphoinositide 3-kinase (PI3K)/AKT signaling increased cell viability. In summary, Rg4 inhibited inflammation and exhibited a protective effect against CLP-induced sepsis, thereby reinforcing cell survival against septic responses.
Collapse
Affiliation(s)
- Go Oun Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Korea
| | - Nayeon Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Korea
| | - Gyu Yong Song
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejon 34134, Korea
- Correspondence: (G.Y.S.); (J.-S.B.); Tel.: +82-42-821-5926 (G.Y.S.); +82-53-950-8570 (J.-S.B.); Fax: +82-42-823-6566 (G.Y.S.); +82-53-950-8557 (J.-S.B.)
| | - Jong-Sup Bae
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Korea
- Correspondence: (G.Y.S.); (J.-S.B.); Tel.: +82-42-821-5926 (G.Y.S.); +82-53-950-8570 (J.-S.B.); Fax: +82-42-823-6566 (G.Y.S.); +82-53-950-8557 (J.-S.B.)
| |
Collapse
|
11
|
Yang Y, Wu JJ, Xia J, Wan Y, Xu JF, Zhang L, Liu D, Chen L, Tang F, Ao H, Peng C. Can aloin develop to medicines or healthcare products? Biomed Pharmacother 2022; 153:113421. [DOI: 10.1016/j.biopha.2022.113421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/13/2022] [Accepted: 07/13/2022] [Indexed: 11/02/2022] Open
|
12
|
Xiao J, Chen S, Chen Y, Su J. The potential health benefits of aloin from genus Aloe. Phytother Res 2022; 36:873-890. [PMID: 35040198 DOI: 10.1002/ptr.7371] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 01/04/2023]
Abstract
The Aloe species is known for its medicinal and cosmetic properties. Aloin is an active ingredient found in the leaves of medicinal plants of the genus Aloe. Aloin has attracted considerable interest for its antiinflammatory, anticancer, antibacterial, and antioxidant activities. However, since its clinical application is restricted by its unclear mechanism of action, a deeper understanding of its pharmacological activity is required. This review provides an overview of current pharmacological and toxicological studies published in English from February 2000 to August 2021. Herein, we summarized the sources and potential health benefits of aloin from a clinical application perspective to guide for further studies on the sources of aloin, aimed at efficiently increasing aloin production. Importantly, the function and mechanism of action of aloin remain unclarified. In future research, it is necessary to develop new approaches for studying the pharmacological molecular mechanisms underlying the activity of this compound against various diseases.
Collapse
Affiliation(s)
- Jianbin Xiao
- College of Life Science, Fujian Normal University, Fuzhou, China.,The Public Service Platform for Industrialization Development Technology of Marine Biological Medicine and Products of the State Oceanic Administration, Fujian Key Laboratory of Special Marine Bioresource Sustainable Utilization, Southern Institute of Oceanography, Fujian Normal University, Fuzhou, China
| | - Siyuan Chen
- College of Life Science, Fujian Normal University, Fuzhou, China.,The Public Service Platform for Industrialization Development Technology of Marine Biological Medicine and Products of the State Oceanic Administration, Fujian Key Laboratory of Special Marine Bioresource Sustainable Utilization, Southern Institute of Oceanography, Fujian Normal University, Fuzhou, China
| | - Youqiang Chen
- College of Life Science, Fujian Normal University, Fuzhou, China.,The Public Service Platform for Industrialization Development Technology of Marine Biological Medicine and Products of the State Oceanic Administration, Fujian Key Laboratory of Special Marine Bioresource Sustainable Utilization, Southern Institute of Oceanography, Fujian Normal University, Fuzhou, China
| | - Jingqian Su
- College of Life Science, Fujian Normal University, Fuzhou, China.,Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| |
Collapse
|
13
|
de Souza Collares Maia Castelo-Branco D, Dos Santos Araújo G, Fonseca XMQC, de Melo Guedes GM, da Rocha MG, Brilhante RSN, de Aguiar Cordeiro R, Sidrim JJC, Pereira-Neto WA, Rocha MFG. Anthraquinones from Aloe spp. inhibit Cryptococcus neoformans sensu stricto: effects against growing and mature biofilms. BIOFOULING 2021; 37:809-817. [PMID: 34634964 DOI: 10.1080/08927014.2021.1958793] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 06/13/2023]
Abstract
This study aimed to evaluate the in vitro effect of aloe emodin, barbaloin and chrysophanol on growing and mature biofilms of Cryptococcus neoformans sensu stricto. The compounds were added at the moment of inducing biofilm growth or after growth for 72 h to evaluate their effects on growing and mature biofilms, respectively. Then, biofilm biomass was evaluated by crystal violet staining and metabolic activity by the XTT reduction assay. Morphological alterations were also evaluated by laser scanning confocal microscopy. Aloe emodin and barbaloin affected growing biofilms and disrupted mature biofilms, reducing metabolic activity by > 60% and biomass by > 70%. Chrysophanol only inhibited mature biofilms, but to a lesser extent. In conclusion, anthraquinones, especially aloe emodin and barbaloin, show a relevant effect against growing and mature biofilms of C. neoformans sensu stricto.
Collapse
Affiliation(s)
- Débora de Souza Collares Maia Castelo-Branco
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Géssica Dos Santos Araújo
- Postgraduate Program in Veterinary Sciences, School of Veterinary, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Xhaulla Maria Quariguasi Cunha Fonseca
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Glaucia Morgana de Melo Guedes
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Maria Gleiciane da Rocha
- Postgraduate Program in Veterinary Sciences, School of Veterinary, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Raimunda Sâmia Nogueira Brilhante
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Rossana de Aguiar Cordeiro
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - José Júlio Costa Sidrim
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Waldemiro Aquino Pereira-Neto
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Marcos Fábio Gadelha Rocha
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
- Postgraduate Program in Veterinary Sciences, School of Veterinary, State University of Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
14
|
Svitina H, Hamman JH, Gouws C. Molecular mechanisms and associated cell signalling pathways underlying the anticancer properties of phytochemical compounds from Aloe species (Review). Exp Ther Med 2021; 22:852. [PMID: 34178125 PMCID: PMC8220653 DOI: 10.3892/etm.2021.10284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/07/2021] [Indexed: 12/14/2022] Open
Abstract
Naturally occurring components from various species of Aloe have been used as traditional folk medicine since the ancient times. Over the last few decades, the therapeutic effects of extracts and phytochemical compounds obtained from Aloe vera have been proven in preclinical and clinical studies. Recently, compounds from other Aloe species apart from Aloe vera have been investigated for the treatment of different diseases, with a particular focus on cancer. In the present review, the effects of phytochemical compounds obtained from different Aloe species are discussed, with a specific focus on the effects on cell signalling in cancer and normal cells, and their selectivity and efficacy. This information will be useful for the application of Aloe-derived compounds as therapeutic agents, either alone or in combination with other standard drugs for cancer treatment.
Collapse
Affiliation(s)
- Hanna Svitina
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™), North-West University, Potchefstroom, North West 2520, South Africa.,Department of Functional Genomics, Institute of Molecular Biology and Genetics of NASU, Kyiv 03143, Ukraine
| | - Josias H Hamman
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™), North-West University, Potchefstroom, North West 2520, South Africa
| | - Chrisna Gouws
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™), North-West University, Potchefstroom, North West 2520, South Africa
| |
Collapse
|
15
|
Liang J, Cui L, Li J, Guan S, Zhang K, Li J. Aloe vera: A Medicinal Plant Used in Skin Wound Healing. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:455-474. [PMID: 33066720 DOI: 10.1089/ten.teb.2020.0236] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skin injury is a major problem threatening human physical and mental health, and how to promote wound healing has been the focus. Developing new wound dressings is an important strategy in skin regeneration. Aloe vera is a medicinal plant with a long history, complex constituents, and various pharmacological activities. Many studies have shown that A. vera plays an important role in promoting wound healing. Adding A. vera to wound dressing has become an ideal way. This review will describe the process of skin injury and wound healing and analyze the role of A. vera in wound healing. In addition, the types of wound dressing and the applications of A. vera in wound dressing will be discussed.
Collapse
Affiliation(s)
- Jiaheng Liang
- School of Life Science, Zhengzhou University, Zhengzhou, P.R. China
| | - Longlong Cui
- School of Life Science, Zhengzhou University, Zhengzhou, P.R. China
| | - Jiankang Li
- School of Life Science, Zhengzhou University, Zhengzhou, P.R. China
| | - Shuaimeng Guan
- School of Life Science, Zhengzhou University, Zhengzhou, P.R. China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou, P.R. China
| | - Jingan Li
- School of Materials Science and Engineering and Henan Key Laboratory of Advanced Magnesium Alloy, Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|