1
|
Li J, Meng ZY, Wen H, Lu CH, Qin Y, Xie YM, Chen Q, Lv JH, Huang F, Zeng ZY. β-sitosterol alleviates pulmonary arterial hypertension by altering smooth muscle cell phenotype and DNA damage/cGAS/STING signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156030. [PMID: 39265206 DOI: 10.1016/j.phymed.2024.156030] [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: 03/13/2024] [Revised: 07/20/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Pulmonary arterial smooth muscle cells (PASMCs) have a neoplastic phenotype characterized by hyperproliferative and anti-apoptotic features that contribute to pulmonary hypertension (PH) development. DNA-sensing adapter protein stimulator of interferon genes (STING) regulate the phenotypic switch of vessel smooth muscle cells. β-sitosterol (SITO) is a nutrient derived from plants that inhibits vascular smooth muscle cell proliferation without notable toxicity. However, the effect of SITO on cancer-like PH-associated pulmonary vascular remodeling and the specific mechanism has not yet be studied. PURPOSE This study investigated the in vitro and in vivo effects of SITO against PH, and its underlying mechanisms. METHODS The therapeutic efficacy of SITO was assessed, and its underlying mechanisms were explored in hypoxia-induced and platelet-derived growth factor (PDGF)-BB-stimulated primary PASMCs and in a monocrotaline (MCT)-induced preclinical PH rat model. SITO or sildenafil (SID) were administered after the MCT intraperitoneal injection. Pulmonary parameters, right heart function, morphology, and PASMCs were cultured for verification. The expression levels of DNA damage/cyclic GMP-AMP synthase (cGAS)/STING were determined using immunofluorescence and Western blotting. STING agonists that interfere with PASMCs were used to determine whether STING mediates the effects of SITO. RESULTS SITO prevented PASMCs proliferation, promoted apoptosis and suppressed phenotypic switching in a dose-dependent manner in vitro and in vivo. In vivo results in rats demonstrated that four weeks of intragastric SITO administration effectively mitigated the MCT-induced elevation of hemodynamic parameters, improved right cardiac function, and reduced pulmonary arteries remodeling. Mechanistically, DNA damage and cGAS/STING/nuclear factor kappa-B signaling activation were observed in rats with PH and cultured PASMCs. SITO exhibited protective effects by suppressing the DNA damage, potentially via inhibiting the expression level of the cGAS/STING signaling pathway. Pharmacological overexpression of STING abolished the anti-proliferative effects of SITO treatment in hypoxia-induced and PDGF-stimulated PASMCs by downregulating PCNA. CONCLUSION SITO may be an attractive agent for PH vascular remodeling by inhibiting proliferation and modulating the phenotypic switch in PASMCs via the DNA damage/cGAS/STING signaling pathway. This study provides a novel therapeutic agent and mediator of the pathological development of PASMCs and PH.
Collapse
Affiliation(s)
- Jing Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China; Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China
| | - Zhong-Yuan Meng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China; Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China
| | - Hong Wen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China; Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China
| | - Chuang-Hong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China; Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China
| | - Yue Qin
- Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China
| | - Yi-Ming Xie
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China; Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China
| | - Qi Chen
- Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China
| | - Jin-Hui Lv
- Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China; Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China.
| | - Zhi-Yu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China; Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, Guangxi 530021, PR China.
| |
Collapse
|
2
|
Xiong L, Chen Q, Liu H. Network pharmacology and molecular docking identified IL-6 as a critical target of Qing Yan He Ji against COVID-19. Medicine (Baltimore) 2024; 103:e40720. [PMID: 39612422 DOI: 10.1097/md.0000000000040720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2024] Open
Abstract
Since the coronavirus disease 2019 (COVID-19) outbreak, although have controlled, severe acute respiratory syndrome coronavirus 2 is constantly mutating and affects people's health. FDA has approved Paxlovid and Molnupiravir for COVID-19 treatment, however, they have not been approved for children under 12 years old. Therefore, it is urgent to explore new drugs for treating COVID-19 in children. As a traditional Chinese medicine, Qing Yan He Ji (QYHJ) has been widely used as an antiviral in our hospital. Therefore, we presumed that it may be ideal for treating COVID-19 and explored its therapeutic effect in patients with COVID-19. The targets and underlying mechanisms of QYHJ against COVID-19 in children were investigated using bioinformatics. QYHJ target sets, and related target genes of COVID-19 were retrieved from public databases. Subsequently, gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed to investigate the potential mechanism of QYHJ against COVID-19. Finally, molecular docking was carried out to analyze the affinity between the effective molecule and the target protein. A total of 15 bioactive ingredients of QYHJ and 111 predicted potential targets of QYHJ against COVID-19 were screened. A protein-protein interaction network and subnetworks identified 21 core target genes. Gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis indicated that QYHJ functions against COVID-19 primarily through antiviral and anti-inflammatory effects. Molecular docking of interleukin-6 (IL-6) revealed that 5 active compounds had relatively stable binding activities with IL-6. Molecular dynamics simulation was performed for molecular docking results, showing IL-6-(4aS,6aR,6aS,6bR,8aR,10R,12aR,14bS)-10-hydroxy-2,2,6a,6b,9,9,12a-heptamethyl-1,3,4,5,6,6a,7,8,8a,10,11,12,13,14b-tetradecahydropicene-4a-carboxylic acid (4aS) complex, IL-6-stigmasterol complex, IL-6-poriferasterol complex, IL-6-sitosterol complex, and IL-6-beta-sitosterol complex had relatively good binding stability. In conclusion, the multi-component and multi-target intervention of QYHJ against COVID-19 is closely related to antiviral and anti-inflammatory activities, which provides a theoretical basis for clinical application.
Collapse
Affiliation(s)
- Lijuan Xiong
- Department of Emergency, Jiangxi Provincial Children's Hospital, the Affiliated Children's Hospital of Nanchang Medical College, Nanchang, China
| | - Qiang Chen
- Jiangxi Pediatric Infectious Diseases Clinical Medical Research Center, Jiangxi Provincial Children's Hospital, the Affiliated Children's Hospital of Nanchang Medical College, Nanchang, China
| | - Hong Liu
- Department of Emergency, Jiangxi Provincial Children's Hospital, the Affiliated Children's Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
3
|
Zhu J, Wang Z, Liu C, Shi M, Guo Z, Li Y, Yu R, Wei J. Study on the Anti-Atherosclerotic Mechanisms of Xin-Tong-Tai Granule Through Network Pharmacology, Molecular Docking, and Experimental Validation. J Inflamm Res 2024; 17:8147-8164. [PMID: 39525320 PMCID: PMC11545721 DOI: 10.2147/jir.s490815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background Xin-Tong-Tai Granule (XTTG), a Chinese medicine (CM) formula, has demonstrated significant therapeutic effects on atherosclerosis (AS) in both clinical and experimental settings. Nonetheless, the mechanisms underlying XTTG's efficacy remain largely unexplored. This study aimed to elucidate the mechanisms through which XTTG acts against AS, employing network pharmacology, molecular docking, and experimental validation techniques. Methods Initially, target identification for the main chemical components of XTTG was conducted using database, followed by determining the intersection targets between these compounds and disease. Protein-protein interaction (PPI) network analysis, Gene Ontology (GO) enrichment, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were subsequently utilized to investigate the potential pathways through which XTTG exerts its effects on AS. Molecular docking was done to confirm the binding efficacy of XTTG's active components. Additionally, the effects of XTTG were evaluated in vitro using oxidized low-density lipoprotein (ox-LDL) induced human aortic vascular smooth muscle cells (HAVSMCs) and in vivo in apolipoprotein E gene knockout (ApoE-/-) mice fed a high-fat diet (HFD). Results 229 therapeutic targets were screened for PPI network and enrichment analysis. Notably, the nuclear factor kappa-B (NF-κB) signaling pathway, along with processes related to inflammation and autophagy, were significantly enriched, highlighting their importance. In vitro studies showed that XTTG repressed cell proliferation and lipid droplet aggregation in ox-LDL-induced HAVSMCs. It also decreased the ratio of phosphorylated NF-κB p65/ NF-κB p65, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels, and elevated microtubule-associated protein light chain 3 (LC3) and decreased p62 protein expression. In vivo, XTTG ameliorated blood lipid profiles and aortic pathology in HFD-fed ApoE-/- mice, reduced NF-κB p65 expression and serum levels of TNF-α and IL-6, increased the ratio of LC3II/LC3I while decreasing p62 protein expression. Conclusion XTTG mitigates AS primarily through anti-inflammatory and autophagy-modulating mechanisms, particularly via inhibition of NF-κB p65 expression. These findings underscore the potential of CM in treating AS and support its further clinical exploration.
Collapse
Affiliation(s)
- Junping Zhu
- School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of TCM Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Ziyan Wang
- First Hospital and First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan Universities of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Chengxin Liu
- First Hospital and First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan Universities of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Min Shi
- School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan Universities of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Zhihua Guo
- School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- First Hospital and First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan Universities of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Ya Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Rong Yu
- School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of TCM Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Jiaming Wei
- School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan Universities of Chinese Medicine, Changsha, 410208, People’s Republic of China
| |
Collapse
|
4
|
Hailati S, Han MY, Dilimulati D, Nueraihemaiti N, Baishan A, Aikebaier A, Zhou WT. Searching for Hub Genes of Quince-Basil Co-Administration Against Atherosclerosis Using Bioinformatics Analysis and Experimental Validation. Pharmaceuticals (Basel) 2024; 17:1433. [PMID: 39598345 PMCID: PMC11597616 DOI: 10.3390/ph17111433] [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: 09/30/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Atherosclerosis (AS) has one of the highest rates of morbidity and death globally. Cydonia oblonga Mill. (quince, COM) and Ocimum basilicum L. (basil, OB) are Uyghur medicines that are often used for anti-inflammatory, anti-tumor, and cardiovascular disease treatment. This study aimed to uncover the hub genes of the quince-basil co-administration against AS and validate them. METHODS Network pharmacology analysis and bioinformatics analysis methods were utilized to map the network and obtain four hub genes. Experiments were performed in vivo and in vitro using HUVEC and zebrafish to validate the therapeutic effect of COM-OB co-administration against AS. Finally, the hub genes were validated by Western blot. RESULTS Screening by network pharmacology analysis and bioinformatics analysis obtained a total of 3302 drug targets, 1963 disease targets, and 1630 DEGs. A series of bioinformatic analyses were utilized to ultimately screen four hub genes, and the stability was also verified by molecular docking and molecular dynamics. COM-OB total flavonoids co-administration significantly decreased PA-induced lipid deposition in HUVEC and reduced high cholesterol-induced fat accumulation in zebrafish. Western blot results showed that COM-OB co-administration significantly affected the expression of hub genes. CONCLUSIONS The study identified and validated four hub genes, COL1A1, COL3A1, BGLAP, and NOX4, thus providing a rationale for the treatment of AS with COM and OB co-administration.
Collapse
Affiliation(s)
- Sendaer Hailati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (S.H.); (M.-Y.H.); (D.D.); (N.N.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Meng-Yuan Han
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (S.H.); (M.-Y.H.); (D.D.); (N.N.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Dilihuma Dilimulati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (S.H.); (M.-Y.H.); (D.D.); (N.N.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Nuerbiye Nueraihemaiti
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (S.H.); (M.-Y.H.); (D.D.); (N.N.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Alhar Baishan
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (S.H.); (M.-Y.H.); (D.D.); (N.N.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Alifeiye Aikebaier
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (S.H.); (M.-Y.H.); (D.D.); (N.N.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Wen-Ting Zhou
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (S.H.); (M.-Y.H.); (D.D.); (N.N.); (A.B.); (A.A.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| |
Collapse
|
5
|
Back P, Yu M, Modaresahmadi S, Hajimirzaei S, Zhang Q, Islam MR, Schwendeman AA, La-Beck NM. Immune Implications of Cholesterol-Containing Lipid Nanoparticles. ACS NANO 2024; 18:28480-28501. [PMID: 39388645 PMCID: PMC11505898 DOI: 10.1021/acsnano.4c06369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024]
Abstract
The majority of clinically approved nanoparticle-mediated therapeutics are lipid nanoparticles (LNPs), and most of these LNPs are liposomes containing cholesterol. LNP formulations significantly alter the drug pharmacokinetics (PK) due to the propensity of nanoparticles for uptake by macrophages. In addition to readily engulfing LNPs, the high expression of cholesterol hydroxylases and reactive oxygen species (ROS) in macrophages suggests that they will readily produce oxysterols from LNP-associated cholesterol. Oxysterols are a heterogeneous group of cholesterol oxidation products that have potent immune modulatory effects. Oxysterols are implicated in the pathogenesis of atherosclerosis and certain malignancies; they have also been found in commercial liposome preparations. Yet, the in vivo metabolic fate of LNP-associated cholesterol remains unclear. We review herein the mechanisms of cellular uptake, trafficking, metabolism, and immune modulation of endogenous nanometer-sized cholesterol particles (i.e., lipoproteins) that are also relevant for cholesterol-containing nanoparticles. We believe that it would be imperative to better understand the in vivo metabolic fate of LNP-associated cholesterol and the immune implications for LNP-therapeutics. We highlight critical knowledge gaps that we believe need to be addressed in order to develop safer and more efficacious lipid nanoparticle delivery systems.
Collapse
Affiliation(s)
- Patricia
Ines Back
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Minzhi Yu
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
| | - Shadan Modaresahmadi
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Sahelosadat Hajimirzaei
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Qisheng Zhang
- Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Md Rakibul Islam
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Anna A. Schwendeman
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
- Biointerfaces
Institute, University of Michigan, North
Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
| | - Ninh M. La-Beck
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
- Department
of Pharmacy Practice, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, Texas 79601, United States
| |
Collapse
|
6
|
Yang S, Zhang Y, Zheng C. β-Sitosterol Mitigates Apoptosis, Oxidative Stress and Inflammatory Response by Inactivating TLR4/NF-кB Pathway in Cell Models of Diabetic Nephropathy. Cell Biochem Biophys 2024:10.1007/s12013-024-01559-4. [PMID: 39424766 DOI: 10.1007/s12013-024-01559-4] [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] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
Podocyte injury plays a pivotal role in the pathogenesis of diabetic nephropathy (DN), leading to proteinuria formation. β-Sitosterol is a natural compound with anti-inflammatory, anti-diabetic, nephroprotective and antioxidant properties. The studyaimed to explore whether and how β-Sitosterol protected podocytes against high glucose (HG)-induced inflammatory andoxidative injury. DN cell models were established by stimulating podocytes or renal tubular epithelial cells (HK-2) cells with 25 mM glucose. Cell viability and apoptosis were evaluated using cell counting kit-8 assays and flow cytometry analyses. Westernblotting was used to quantify protein levels of genes related to podocyte injury, HK-2 cell damage, inflammation, and TLR4/NF-кB pathway. Contents of oxidative stress biomarkers were evaluated by corresponding commercial kits while proinflammatorycytokine levels were determined by enzyme-linked immunosorbent assay. Immunofluorescence staining was performed todetect intracellular levels of reactive oxygen species (ROS) and Nrf2 nuclear translocation. Experimental results revealed that HG treatment induced podocyte dysfunction by impairing cell viability while accelerating theapoptosis, and the changes were reversed by β-sitosterol treatment. Moreover, β-sitosterol repressed HG-evoked oxidative stressby reducing ROS and malondialdehyde (MDA) levels while increasing activities of antioxidant enzymes. The reduction ofproinflammatory cytokines mediated by β-sitosterol in HG-stimulated podocytes suggested the anti-inflammatory role of β-sitosterol. Additionally, the activation of the TLR4/NF-кB signaling induced by HG was inhibited by β-sitosterol in podocytes.Inactivation of the TLR4 using TAK-242 enhanced the protective effects of β-sitosterol against HG-mediated oxidative stressand inflammation. Similarly, β-sitosterol also protected HK-2 cells from HG-induced oxidative stress, inflammation, andapoptosis. In summary, β-sitosterol exerts anti-inflammatory, anti-oxidative, and anti-apoptotic activities in HG-induced podocytes or HK-2 cells by inhibiting TLR4/NF-кB signaling.
Collapse
Affiliation(s)
| | | | - Chenghong Zheng
- Hubei University of Chinese Medicine, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China.
| |
Collapse
|
7
|
Li M, Meng Y, Hong X, Chai H, Huang J, Wang F, Zhang W, Wang J, Liu Q, Xu Y. Anti-atherosclerotic effect of tetrahydroxy stilbene glucoside via dual-targeting of hepatic lipid metabolisms and aortic M2 macrophage polarization in ApoE -/- mice. J Pharm Biomed Anal 2024; 248:116338. [PMID: 38971092 DOI: 10.1016/j.jpba.2024.116338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 05/13/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Tetrahydroxy stilbene glucoside (TSG) is a water-soluble natural product that has shown potential in treating atherosclerosis (AS). However, its underlying mechanisms remain unclear. Here, we demonstrate that an 8-week TSG treatment (100 mg/kg/d) significantly reduces atherosclerotic lesions and alleviates dyslipidemia symptoms in ApoE-/- mice. 1H nuclear magnetic resonance metabolomic analysis reveals differences in both lipid components and water-soluble metabolites in the livers of AS mice compared to control groups, and TSG treatment shifts the metabolic profiles of AS mice towards a normal state. At the transcriptional level, TSG significantly restores the expression of fatty acid metabolism-related genes (Srepb-1c, Fasn, Scd1, Gpat1, Dgat1, Pparα and Cpt1α), and regulates the expression levels of disturbed cholesterol metabolism-related genes (Srebp2, Hmgcr, Ldlr, Acat1, Acat2 and Cyp7a1) associated with lipid metabolism. Furthermore, at the cellular level, TSG remarkably polarizes aortic macrophages to their M2 phenotype. Our data demonstrate that TSG alleviates arthrosclerosis by dual-targeting to hepatic lipid metabolism and aortic M2 macrophage polarization in ApoE-/- mice, with significant implications for translational medicine and the treatment of AS using natural products.
Collapse
Affiliation(s)
- Minghui Li
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Yuanyuan Meng
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Xuelian Hong
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Hui Chai
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Jianye Huang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Fengge Wang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Wenjie Zhang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Juncheng Wang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Qingwang Liu
- Institute of Heath & Medical Technology, Hefei Institute of Physical Science, Chinese Academy of Sciences, Hefei 230031, China.
| | - Yuekang Xu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China.
| |
Collapse
|
8
|
Jiang S, Gao K, Zhang F, Wang Y, He X, Yang J. β-sitosterol alleviates atherosclerosis by regulating catalase. Heliyon 2024; 10:e35639. [PMID: 39165938 PMCID: PMC11334795 DOI: 10.1016/j.heliyon.2024.e35639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024] Open
Abstract
The aim of this study is to investigate the main active components of Gegen (Puerariae Lobatae Radix) on atherosclerosis and its mechanism of action. Bioinformatics analysis showed that β-sitosterol was the most likely active ingredient to mediate the anti-atherosclerotic effects. In vivo experiments showed that β-sitosterol inhibited plaque formation and platelet activation, and decreased serum total cholesterol (TC) and triglyceride (TG) levels. In vitro experiments showed that β-sitosterol can inhibit lipid deposition and phenotypic transformation of vascular smooth muscle cells (VSMCs). However, knocking down catalase (CAT), the direct target of β-sitosterol, not only promoted lipid deposition and phenotypic transformation of VSMCs, but also activated the PI3K/Akt/mTOR pathway, and the mTOR inhibitor (ink-128) can eliminate the effect of CAT knockdown, suggesting that β-sitosterol may inhibit lipid deposition and phenotypic transformation of VSMCs by activating CAT and silencing the PI3K/Akt/mTOR signaling pathway, thereby alleviating atherosclerosis.
Collapse
Affiliation(s)
- Shuntao Jiang
- Department of Cardiovascular Medicine, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi, 563000, Guizhou, China
| | - Kui Gao
- Department of Cardiology, People's Hospital of Lanshan District, NO.566, Lanshan West Road, Lanshan District, Rizhao, 276800, Shandong, China
| | - Furong Zhang
- Department of Cardiology, Huantai People's Hospital, No.2198, Huan Tai Avenue, Huantai suo Town, Zibo, 256400, Shandong, China
| | - Yanli Wang
- Department of Geriatrics, Traditional Chinese and Western Medicine Hospital of Qingdao, No.3, Jiaxiang Road, Qingdao, 266000, Shandong, China
| | - Xiaojing He
- Department of Cardiology, The First People's Hospital of Ningyang, No.872, Jinyang Street, Ningyang County, Taian, 271400, Shandong, China
| | - Jun Yang
- Department of Outpatient, The First People's Hospital of Yunnan, No.157, Jinbi road, Kunming, 650000, Yunnan, China
| |
Collapse
|
9
|
Ai J, Tang X, Mao B, Zhang Q, Zhao J, Chen W, Cui S. Gut microbiota: a superior operator for dietary phytochemicals to improve atherosclerosis. Crit Rev Food Sci Nutr 2024:1-23. [PMID: 38940319 DOI: 10.1080/10408398.2024.2369169] [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: 06/29/2024]
Abstract
Mounting evidence implicates the gut microbiota as a possible key susceptibility factor for atherosclerosis (AS). The employment of dietary phytochemicals that strive to target the gut microbiota has gained scientific support for treating AS. This study conducted a general overview of the links between the gut microbiota and AS, and summarized available evidence that dietary phytochemicals improve AS via manipulating gut microbiota. Then, the microbial metabolism of several dietary phytochemicals was summarized, along with a discussion on the metabolites formed and the biotransformation pathways involving key gut bacteria and enzymes. This study additionally focused on the anti-atherosclerotic potential of representative metabolites from dietary phytochemicals, and investigated their underlying molecular mechanisms. In summary, microbiota-dependent dietary phytochemical therapy is a promising strategy for AS management, and knowledge of "phytochemical-microbiota-biotransformation" may be a breakthrough in the search for novel anti-atherogenic agents.
Collapse
Affiliation(s)
- Jian Ai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xin Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
10
|
Duarte RDC, Iannetta PPM, Gomes AM, Vasconcelos MW. More than a meat- or synthetic nitrogen fertiliser-substitute: a review of legume phytochemicals as drivers of 'One Health' via their influence on the functional diversity of soil- and gut-microbes. FRONTIERS IN PLANT SCIENCE 2024; 15:1337653. [PMID: 38450400 PMCID: PMC10915056 DOI: 10.3389/fpls.2024.1337653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/30/2024] [Indexed: 03/08/2024]
Abstract
Legumes are essential to healthy agroecosystems, with a rich phytochemical content that impacts overall human and animal well-being and environmental sustainability. While these phytochemicals can have both positive and negative effects, legumes have traditionally been bred to produce genotypes with lower levels of certain plant phytochemicals, specifically those commonly termed as 'antifeedants' including phenolic compounds, saponins, alkaloids, tannins, and raffinose family oligosaccharides (RFOs). However, when incorporated into a balanced diet, such legume phytochemicals can offer health benefits for both humans and animals. They can positively influence the human gut microbiome by promoting the growth of beneficial bacteria, contributing to gut health, and demonstrating anti-inflammatory and antioxidant properties. Beyond their nutritional value, legume phytochemicals also play a vital role in soil health. The phytochemical containing residues from their shoots and roots usually remain in-field to positively affect soil nutrient status and microbiome diversity, so enhancing soil functions and benefiting performance and yield of following crops. This review explores the role of legume phytochemicals from a 'one health' perspective, examining their on soil- and gut-microbial ecology, bridging the gap between human nutrition and agroecological science.
Collapse
Affiliation(s)
- Rafael D. C. Duarte
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Pietro P. M. Iannetta
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
- Ecological Sciences, James Hutton Institute, Dundee, United Kingdom
| | - Ana M. Gomes
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Marta W. Vasconcelos
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina – Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| |
Collapse
|
11
|
Fan C, Xu J, Tong H, Fang Y, Chen Y, Lin Y, Chen R, Chen F, Wu G. Gut-brain communication mediates the impact of dietary lipids on cognitive capacity. Food Funct 2024; 15:1803-1824. [PMID: 38314832 DOI: 10.1039/d3fo05288e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Cognitive impairment, as a prevalent symptom of nervous system disorders, poses one of the most challenging aspects in the management of brain diseases. Lipids present in the cell membranes of all neurons within the brain and dietary lipids can regulate the cognition and memory function. In recent years, the advancements in gut microbiome research have enabled the exploration of dietary lipids targeting the gut-brain axis as a strategy for regulating cognition. This present review provides an in-depth overview of how lipids modulate cognition via the gut-brain axis depending on metabolic, immune, neural and endocrine pathways. It also comprehensively analyzes the effects of diverse lipids on the gut microbiota and intestinal barrier function, thereby affecting the central nervous system and cognitive capacity. Moreover, comparative analysis of the positive and negative effects is presented between beneficial and detrimental lipids. The former encompass monounsaturated fatty acids, short-chain fatty acids, omega-3 polyunsaturated fatty acids, phospholipids, phytosterols, fungal sterols and bioactive lipid-soluble vitamins, as well as lipid-derived gut metabolites, whereas the latter (detrimental lipids) include medium- or long-chain fatty acids, excessive proportions of n-6 polyunsaturated fatty acids, industrial trans fatty acids, and zoosterols. To sum up, the focus of this review is on how gut-brain communication mediates the impact of dietary lipids on cognitive capacity, providing a novel theoretical foundation for promoting brain cognitive health and scientific lipid consumption patterns.
Collapse
Affiliation(s)
- Chenhan Fan
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Jingxuan Xu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Haoxiang Tong
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Yucheng Fang
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Yiming Chen
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Yangzhuo Lin
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Rui Chen
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Fuhao Chen
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Guoqing Wu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
12
|
Nandi S, Nag A, Khatua S, Sen S, Chakraborty N, Naskar A, Acharya K, Calina D, Sharifi-Rad J. Anticancer activity and other biomedical properties of β-sitosterol: Bridging phytochemistry and current pharmacological evidence for future translational approaches. Phytother Res 2024; 38:592-619. [PMID: 37929761 DOI: 10.1002/ptr.8061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 11/07/2023]
Abstract
Sterols, including β-sitosterol, are essential components of cellular membranes in both plant and animal cells. Despite being a major phytosterol in various plant materials, comprehensive scientific knowledge regarding the properties of β-sitosterol and its potential applications is essential for scholarly pursuits and utilization purposes. β-sitosterol shares similar chemical characteristics with cholesterol and exhibits several pharmacological activities without major toxicity. This study aims to bridge the gap between phytochemistry and current pharmacological evidence of β-sitosterol, focusing on its anticancer activity and other biomedical properties. The goal is to provide a comprehensive understanding of β-sitosterol's potential for future translational approaches. A thorough examination of the literature was conducted to gather relevant information on the biological properties of β-sitosterol, particularly its anticancer therapeutic potential. Various databases were searched, including PubMed/MedLine, Scopus, Google Scholar, and Web of Science using appropriate keywords. Studies investigating the effects of β-sitosterol on different types of cancer were analyzed, focusing on mechanisms of action, pharmacological screening, and chemosensitizing properties. Modern pharmacological screening studies have revealed the potential anticancer therapeutic properties of β-sitosterol against various types of cancer, including leukemia, lung, stomach, breast, colon, ovarian, and prostate cancer. β-sitosterol has demonstrated chemosensitizing effects on cancer cells, interfering with multiple cell signaling pathways involved in proliferation, cell cycle arrest, apoptosis, survival, metastasis invasion, angiogenesis, and inflammation. Structural derivatives of β-sitosterol have also shown anti-cancer effects. However, research in the field of drug delivery and the detailed mode of action of β-sitosterol-mediated anticancer activities remains limited. β-sitosterol, as a non-toxic compound with significant pharmacological potential, exhibits promising anticancer effects against various cancer types. Despite being relatively less potent than conventional cancer chemotherapeutics, β-sitosterol holds potential as a safe and effective nutraceutical against cancer. Further comprehensive studies are recommended to explore the biological properties of β-sitosterol, including its mode of action, and develop novel formulations for its potential use in cancer treatment. This review provides a foundation for future investigations and highlights the need for further research on β-sitosterol as a potent superfood in combating cancer.
Collapse
Affiliation(s)
- Sudeshna Nandi
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata, India
| | - Anish Nag
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, India
| | - Somanjana Khatua
- Department of Botany, Faculty of Science, University of Allahabad, Prayagraj, India
| | - Surjit Sen
- Department of Botany, Fakir Chand College, Kolkata, India
| | | | - Arghya Naskar
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata, India
| | - Krishnendu Acharya
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata, India
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | | |
Collapse
|
13
|
Li S, Chen J, Liu Y, Qiu H, Gao W, Che K, Zhou B, Liu R, Hu W. Preparation of Citral Oleogel and Antimicrobial Properties. Gels 2023; 9:930. [PMID: 38131916 PMCID: PMC10742588 DOI: 10.3390/gels9120930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
The objective of this study was to analyze a natural and safe oleogel with antimicrobial properties that can replace animal fats while lengthening the product's shelf life. The oleogel was created using direct dispersion (MG-SO), and its material characterization exhibited the exceptional performance of the hybrid gelant. Additionally, citral was integrated into the oil gel to prepare the citral oleogel (MG-SO). The antimicrobial nature of the material was examined and the findings revealed that it inhibited the growth of various experimental model bacteria, including Escherichia coli, Staphylococcus aureus, Aspergillus niger, Botrytis cinerea, and Rhizopus stolonifer. In addition, the material had a comparable inhibitory impact on airborne microorganisms. Lastly, MG-SON was utilized in plant-based meat patties and demonstrated an ability to significantly reduce the growth rate of microorganisms.
Collapse
Affiliation(s)
- Shangjian Li
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519041, China
- College of Life Science, Jilin University, Changchun 130015, China
| | - Jiajia Chen
- Zhuhai Lizhu Microsphere Technology Co., Zhuhai 519000, China
| | - Yuntong Liu
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519041, China
- College of Life Science, Jilin University, Changchun 130015, China
| | - Honghao Qiu
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519041, China
- College of Life Science, Jilin University, Changchun 130015, China
| | - Wei Gao
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519041, China
- College of Life Science, Jilin University, Changchun 130015, China
| | - Kundian Che
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519041, China
- College of Life Science, Jilin University, Changchun 130015, China
| | - Baogang Zhou
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519041, China
- College of Life Science, Jilin University, Changchun 130015, China
| | - Ran Liu
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519041, China
- College of Life Science, Jilin University, Changchun 130015, China
| | - Wenzhong Hu
- School of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai 519041, China
- College of Life Science, Dalian Minzu University, Dalian 116600, China
| |
Collapse
|
14
|
Micek A, Bolesławska I, Jagielski P, Konopka K, Waśkiewicz A, Witkowska AM, Przysławski J, Godos J. Association of dietary intake of polyphenols, lignans, and phytosterols with immune-stimulating microbiota and COVID-19 risk in a group of Polish men and women. Front Nutr 2023; 10:1241016. [PMID: 37599696 PMCID: PMC10436747 DOI: 10.3389/fnut.2023.1241016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
Objectives Devastating consequences of COVID-19 disease enhanced the role of promoting prevention-focused practices. Among targeted efforts, diet is regarded as one of the potential factors which can affect immune function and optimal nutrition is postulated as the method of augmentation of people's viral resistance. As epidemiological evidence is scarce, the present study aimed to explore the association between dietary intake of total polyphenols, lignans and plant sterols and the abundance of immunomodulatory gut microbiota such as Enterococcus spp. and Escherichia coli and the risk of developing COVID-19 disease. Methods Demographic data, dietary habits, physical activity as well as the composition of body and gut microbiota were analyzed in a sample of 95 young healthy individuals. Dietary polyphenol, lignan and plant sterol intakes have been retrieved based on the amount of food consumed by the participants, the phytochemical content was assessed in laboratory analysis and using available databases. Results For all investigated polyphenols and phytosterols, except campesterol, every unit increase in the tertile of intake category was associated with a decrease in the odds of contracting COVID-19. The risk reduction ranged from several dozen percent to 70 %, depending on the individual plant-based chemical, and after controlling for basic covariates it was statistically significant for secoisolariciresinol (OR = 0.28, 95% CI: 0.11-0.61), total phytosterols (OR = 0.47, 95% CI: 0.22-0.95) and for stigmasterols (OR = 0.34, 95% CI: 0.14-0.72). We found an inverse association between increased β-sitosterol intake and phytosterols in total and the occurrence of Escherichia coli in stool samples outside reference values, with 72% (OR = 0.28, 95% CI: 0.08-0.86) and 66% (OR = 0.34, 95% CI: 0.10-1.08) reduced odds of abnormal level of bacteria for the highest compared with the lowest tertile of phytochemical consumption. Additionally, there was a trend of more frequent presence of Enterococcus spp. at relevant level in people with a higher intake of lariciresinol. Conclusion The beneficial effects of polyphenols and phytosterols should be emphasized and these plant-based compounds should be regarded in the context of their utility as antiviral agents preventing influenza-type infections.
Collapse
Affiliation(s)
- Agnieszka Micek
- Statistical Laboratory, Jagiellonian University Medical College, Cracow, Poland
| | - Izabela Bolesławska
- Department of Bromatology, Poznan University of Medical Sciences, Poznań, Poland
| | - Paweł Jagielski
- Department of Nutrition and Drug Research, Faculty of Health Sciences, Institute of Public Health, Jagiellonian University Medical College, Kraków, Poland
| | - Kamil Konopka
- Department of Oncology, Jagiellonian University Medical College, Kraków, Poland
| | - Anna Waśkiewicz
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, National Institute of Cardiology, Warszawa, Poland
| | - Anna Maria Witkowska
- Department of Food Biotechnology, Medical University of Bialystok, Białystok, Poland
| | - Juliusz Przysławski
- Department of Bromatology, Poznan University of Medical Sciences, Poznań, Poland
| | - Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
15
|
Chen Y, Gan Y, Yu J, Ye X, Yu W. Key ingredients in Verbena officinalis and determination of their anti-atherosclerotic effect using a computer-aided drug design approach. FRONTIERS IN PLANT SCIENCE 2023; 14:1154266. [PMID: 37077636 PMCID: PMC10106644 DOI: 10.3389/fpls.2023.1154266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/16/2023] [Indexed: 05/03/2023]
Abstract
Lipid metabolism disorders may considerably contribute to the formation and development of atherosclerosis (AS). Traditional Chinese medicine has received considerable attention in recent years owing to its ability to treat lipid metabolism disorders using multiple components and targets. Verbena officinalis (VO), a Chinese herbal medicine, exhibits anti-inflammatory, analgesic, immunomodulatory, and neuroprotective effects. Evidence suggests that VO regulates lipid metabolism; however, its role in AS remains unclear. In the present study, an integrated network pharmacology approach, molecular docking, and molecular dynamics simulation (MDS) were applied to examine the mechanism of VO against AS. Analysis revealed 209 potential targets for the 11 main ingredients in VO. Further, 2698 mechanistic targets for AS were identified, including 147 intersection targets between VO and AS. Quercetin, luteolin, and kaempferol were considered key ingredients for the treatment of AS based on a potential ingredient target-AS target network. GO analysis revealed that biological processes were primarily associated with responses to xenobiotic stimuli, cellular responses to lipids, and responses to hormones. Cell components were predominantly focused on the membrane microdomain, membrane raft, and caveola nucleus. Molecular functions were mainly focused on DNA-binding transcription factor binding, RNA polymerase II-specific DNA-binding transcription factor binding, and transcription factor binding. KEGG pathway enrichment analysis identified pathways in cancer, fluid shear stress, and atherosclerosis, with lipid and atherosclerosis being the most significantly enriched pathways. Molecular docking revealed that three key ingredients in VO (i.e., quercetin, luteolin, and kaempferol) strongly interacted with three potential targets (i.e., AKT1, IL-6, and TNF-α). Further, MDS revealed that quercetin had a stronger binding affinity for AKT1. These findings suggest that VO has beneficial effects on AS via these potential targets that are closely related to the lipid and atherosclerosis pathways. Our study utilized a new computer-aided drug design to identify key ingredients, potential targets, various biological processes, and multiple pathways associated with the clinical roles of VO in AS, which provides a comprehensive and systemic pharmacological explanation for the anti-atherosclerotic activity of VO.
Collapse
Affiliation(s)
- Yuting Chen
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Yuanyuan Gan
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Jingxuan Yu
- Clinical Medical College, Changsha Medical University, Changsha, Hunan, China
| | - Xiao Ye
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Wei Yu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning, Hubei, China
- *Correspondence: Wei Yu,
| |
Collapse
|