1
|
Susawaengsup C, Choengpanya K, Sornsakdanuphap J, Tabtimmai L, Chaiharn M, Bhuyar P. Phytochemical and Pharmacological Properties of a Traditional Herb, Strobilanthes Cusia (Nees) Kuntze. Mol Biotechnol 2024; 66:2860-2871. [PMID: 37773314 DOI: 10.1007/s12033-023-00897-7] [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: 07/01/2023] [Accepted: 09/05/2023] [Indexed: 10/01/2023]
Abstract
The present investigation aimed to determine the effectiveness of bioactive components extracted from Hom herbs (Strobilanthes cusia (Nees) Kuntze) using the solvent-free microwave-assisted extraction (MAE) method. The obtained bioactive components were analyzed for total phenolic content (TPC) and active ingredient content. The Hom extracts were examined for antioxidant, antibacterial, anti-inflammatory, cytotoxic, and anticancer activities. The comparative analysis of extraction methods MAE was studied by using different solvents such as ethanol (EtOH), 50% ethanol (50EtOH) and distilled water (DW). The results obtained by the MAE method with DW as solvent show the TPC of 104.41±1.36 mg GAE/g crude and tryptanthrin 0.1138±0.0014 mg/g crude and indigo 0.0622±0.0015 mg/g crude. Comparatively, values detected in the 50% EtOH extract were not significantly different at the 95% confidence level. At the same time, levels of indirubin were detected at levels equivalent to that of ethanol extracts. The DW extract from MAE had an IC50 value against the DPPH scavenging assay of 0.1927±0.0756 mg/ml, comparable to the test results of extracts of ethanol and 50% ethanol. The bioactive extracted using the MAE with water as solvent had minimum inhibitory concentration (MIC) and could suppress infection at 10 mg/disc. It was also observed that the extracts from the conventional extraction technique using ethanol as the solvent continued to be highly effective against Bacillus cereus even after employing the EtOH or 50% EtOH. Hom extract's MIC value representing inhibiting B. cereus was 0.625 mg/disc. Still, EtOH-extracted Hom demonstrated the highest cytotoxicity against 16HBEo- by reducing cell survival rate by less than 50% while the others did not. Interestingly, Hom that had been extracted using 50EtOH and DW with MAE had an anticancer impact on A549 by reducing the survival rate in a dose-dependent manner.
Collapse
Affiliation(s)
- Chanthana Susawaengsup
- Maejo University, Phrae Campus, Mae Sai, Rong Kwang District, Phrae, 54140, Thailand.
- International Industry and Agriculture Innovation Research Center (IIAR), International College, Maejo University, Nongharn, Sansai District, Chiang Mai, 50290, Thailand.
| | - Khuanjarat Choengpanya
- Maejo University, Phrae Campus, Mae Sai, Rong Kwang District, Phrae, 54140, Thailand
- International Industry and Agriculture Innovation Research Center (IIAR), International College, Maejo University, Nongharn, Sansai District, Chiang Mai, 50290, Thailand
| | - Jirapong Sornsakdanuphap
- Maejo University, Phrae Campus, Mae Sai, Rong Kwang District, Phrae, 54140, Thailand
- International Industry and Agriculture Innovation Research Center (IIAR), International College, Maejo University, Nongharn, Sansai District, Chiang Mai, 50290, Thailand
| | - Lueacha Tabtimmai
- Department of Biotechnology, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bang Sue, Bangkok, 10800, Thailand
| | - Mathurot Chaiharn
- Division of Biotechnology, Faculty of Science, Maejo University, Sansai District, Chiang Mai, 50290, Thailand
| | - Prakash Bhuyar
- International Industry and Agriculture Innovation Research Center (IIAR), International College, Maejo University, Nongharn, Sansai District, Chiang Mai, 50290, Thailand.
- International College, Maejo University, Nongharn, Sansai District, Chiang Mai, 50290, Thailand.
| |
Collapse
|
2
|
Kim H, Jeong S, Kim SW, Kim HJ, Kim DY, Yook TH, Yang G. Indigo Naturalis in Inflammatory Bowel Disease: mechanisms of action and insights from clinical trials. J Pharmacopuncture 2024; 27:59-69. [PMID: 38948310 PMCID: PMC11194518 DOI: 10.3831/kpi.2024.27.2.59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/02/2024] [Accepted: 03/20/2024] [Indexed: 07/02/2024] Open
Abstract
This study investigates the therapeutic potential of Indigo Naturalis (IN) in treating a Inflammatory Bowel Disease (IBD). The objective is to comprehensively examine the effects and pharmacological mechanisms of IN on IBD, assessing its potential as an novel treatment for IBD. Analysis of 11 selected papers is conducted to understand the effects of IN, focusing on compounds like indirubin, isatin, indigo, and tryptanthrin. This study evaluates their impact on Disease Activity Index (DAI) score, colon length, mucosal damage, and macrophage infiltration in Dextran Sulfate Sodium (DSS)-induced colitis mice. Additionally, It investigate into the anti-inflammatory mechanisms, including Aryl hydrocarbon Receptor (AhR) pathway activation, Nuclear Factor kappa B (NF-κB)/nod-like receptor family pyrin domain containing 3 (NLRP3)/Interleukin 1 beta (IL-1β) inhibition, and modulation of Toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88 (MYD88)/NF-κB and Mitogen Activated Protein Kinase (MAPK) pathways. Immunomodulatory effects on T helper 17 (Th17)/regulatory T cell (Treg cell) balance and Glycogen synthase kinase-3 beta (GSK3-β) expression are also explored. Furthermore, the study addresses the role of IN in restoring intestinal microbiota diversity, reducing pathogenic bacteria, and increasing beneficial bacteria. The findings reveal that IN, particularly indirubin and indigo, demonstrates significant improvements in DAI score, colon length, mucosal damage, and macrophage infiltration in DSS-induced colitis mice. The anti-inflammatory effects are attributed to the activation of the AhR pathway, inhibition of inflammatory pathways, and modulation of immune responses. These results exhibit the potential of IN in IBD treatment. Notably, the restoration of intestinal microbiota diversity and balance further supports its efficacy. IN emerges as a promising and effective treatment for IBD, demonstrating anti-inflammatory effects and positive outcomes in preclinical studies. However, potential side effects necessitate further investigation for safe therapeutic development. The study underscores the need for future research to explore a broader range of active ingredients in IN to enhance therapeutic efficacy and safety.
Collapse
Affiliation(s)
- Hyeonjin Kim
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Soohyun Jeong
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Sung Wook Kim
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Hyung-Jin Kim
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Dae Yong Kim
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Tae Han Yook
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| | - Gabsik Yang
- Department of Korean Medicine, College of Korea Medicine, Woosuk University, Jeonju, Republic of Korea
| |
Collapse
|
3
|
Hartl A, Polleichtner A, Novak J. "Purplish Blue" or "Greenish Grey"? Indigo Qualities and Extraction Yields from Six Species. PLANTS (BASEL, SWITZERLAND) 2024; 13:918. [PMID: 38611450 PMCID: PMC11013892 DOI: 10.3390/plants13070918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/27/2024] [Accepted: 03/08/2024] [Indexed: 04/14/2024]
Abstract
Indigo quality is determined by its indigotin content. Another quality indicator is colour. For an evaluation of species, indigo samples from Indigofera tinctoria, Indigofera suffruticosa, Indigofera arrecta, Persicaria tinctoria, Strobilanthes cusia and Wrightia laevis cultivated in Austria and China were visually classified and analysed spectrophotometrically and using a L*a*b* measuring device. In addition to a standardised hot-extraction method without lime, some samples were extracted simulating traditional methods at ambient temperatures using lime. The highest indigotin contents were achieved with Indigofera arrecta (55%, Austria) and Strobilanthes cusia (56%, China). There were no statistically significant differences between the indigo extraction yields of the species cultivated in Austria, but Indigofera arrecta and Persicaria tinctoria had statistically significantly higher indigotin extraction yields than Indigofera tinctoria and Indigofera suffruticosa. From the species extracted in China, Strobilanthes cusia showed higher values in all parameters than Indigofera tinctoria, Indigofera suffruticosa and Wrightia laevis. Compared with the standardised method, the method simulating local practice yielded more indigo but had a lower indigotin content; the indigotin extraction yields did not differ greatly. L*a*b* values enabled precise estimations of the indigotin content, making it an interesting option for quality control, as inexpensive, easy-to-handle L*a*b* measuring instruments have become available.
Collapse
Affiliation(s)
- Anna Hartl
- Working Group Knowledge Systems and Innovation, Institute of Organic Farming, Department of Sustainable Agricultural Systems, University of Natural Resources and Life Sciences, Vienna, Gregor-Mendel-Strasse 33, 1180 Vienna, Austria
| | - Andrea Polleichtner
- Working Group Soil Fertility and Cropping Systems, Institute of Organic Farming, Department of Sustainable Agricultural Systems, University of Natural Resources and Life Sciences, Vienna, Gregor-Mendel-Strasse 33, 1180 Vienna, Austria
| | - Johannes Novak
- Institute of Animal Nutrition and Functional Plant Compounds, Department Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Veterinärplatz 1, 1210 Vienna, Austria;
| |
Collapse
|
4
|
Xu X, Taha R, Chu C, Xiao L, Wang T, Wang X, Huang X, Jiang Z, Sun L. Indirubin mediates adverse intestinal reactions in guinea pigs by downregulating the expression of AchE through AhR. Xenobiotica 2024; 54:83-94. [PMID: 38164702 DOI: 10.1080/00498254.2023.2297745] [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: 10/22/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Indirubin is the main component of the traditional Chinese medicine Indigo naturalis (IN), a potent agonist of aryl hydrocarbon receptors (AhRs). In China, IN is used to treat psoriasis and ulcerative colitis, and indirubin is used for the treatment of chronic myelogenous leukaemia. However, IN and indirubin have adverse reactions, such as abdominal pain, diarrhoea, and intussusception, and their specific mechanism is unclear.The purpose of our research was to determine the specific mechanism underlying the adverse effects of IN and indirubin. By tracking the modifications in guinea pigs after the intragastric administration of indirubin for 28 days.The results demonstrate that indirubin could accelerate bowel movements and decrease intestinal acetylcholinesterase (AchE) expression. Experiments with NCM460 cells revealed that indirubin significantly reduced the expression of AchE, and the AchE levels were increased after the silencing of AhR and re-exposure to indirubin.This study showed that the inhibition of AchE expression by indirubin plays a key role in the occurrence of adverse reactions to indirubin and that the underlying mechanism is related to AhR-mediated AchE downregulation.
Collapse
Affiliation(s)
- Xiaoting Xu
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Reham Taha
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Chenghan Chu
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Li Xiao
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou, China
| | - Tao Wang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Xinzhi Wang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Xin Huang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Zhenzhou Jiang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Lixin Sun
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
5
|
Tran HL, Lai KH, Chang HS, Chen YS, Wang HC, Yang SS, Chang HW, Hsu CM, Yen CH, Hsiao HH. Indigofera suffruticosa aerial parts extract induce G2/M arrest and ATR/CHK1 pathway in Jurkat cells. BMC Complement Med Ther 2024; 24:28. [PMID: 38195460 PMCID: PMC10775588 DOI: 10.1186/s12906-023-04325-w] [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: 07/11/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Indigofera suffruticosa Mill. is used as a folk medicine for treating patients with leukemia, however very little is known regarding the molecular mechanism of its anti-leukemic activity and the chemical profile of the active extract. The present study aimed to reveal the molecular effect of I. suffruticosa aerial parts extract (ISAE) on leukemia cells and its chemical constituents. METHODS Cytotoxicity of ISAE were determined by resazurin viability assay, multitox - Glo multiplex cytotoxicity assay, and Annexin V staining assay. Cell cycle profiles were revealed by propidium iodide staining assay. The effects of ISAE on G2/M arrest signaling and DNA damage were evaluated by Western blot assay and phospho-H2A.X staining assay. The chemical profile of ISAE were determined by tandem mass spectroscopy and molecular networking approach. RESULTS We showed that the acute lymphoblastic leukemia cell line Jurkat cell was more responsive to ISAE treatment than other leukemia cell lines. In contrast, ISAE did not induce cytotoxic effects in normal fibroblast cells. Cell cycle analysis revealed that ISAE triggered G2/M arrest in Jurkat cells in dose- and time-dependent manners. Elevation of annexin V-stained cells and caspase 3/7 activity suggested ISAE-induced apoptosis. Furthermore, ISAE alone could increase the phosphorylation of CDK1 at Y15 and activate the ATR/CHK1/Wee1/CDC25C signaling pathway. However, the addition of caffeine, a widely used ATR inhibitor to ISAE, reduced the phosphorylation of ATR, CHK1, and CDK1, as well as G2/M arrest in Jurkat cells. Moreover, increased phospho-H2A.X stained cells indicated the involvement of DNA damage in the anti-leukemic effect of ISAE. Finally, qualitative analysis using UPLC-tandem mass spectroscopy and molecular networking revealed that tryptanthrin was the most abundant organoheterocyclic metabolite in ISAE. At equivalent concentrations to ISAE, tryptanthrin induced G2/M arrest of Jurkat cells, which can be prevented by caffeine. CONCLUSIONS ISAE causes G2/M arrest via activating ATR/CHK1/CDK1 pathway and tryptanthrin is one of the active components of ISAE. Our findings provide subtle support to the traditional use of I. suffruitcosa in leukemia management in folk medicine.
Collapse
Affiliation(s)
- Hong-Loan Tran
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Kuei-Hung Lai
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
| | - Hsun-Shuo Chang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yi-Siao Chen
- Ph.D. Program in Environmental and Occupational Medicine, College of Medicine, Kaohsiung Medical University and National Health Research Institutes, Kaohsiung, 80708, Taiwan
| | - Hui-Chun Wang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Shuen-Shin Yang
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chin-Mu Hsu
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
| | - Chia-Hung Yen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan.
| | - Hui-Hua Hsiao
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan.
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan.
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
6
|
Frumkin LR. Letter by Frumkin Regarding Article, "Dramatically Improved Severe Pulmonary Arterial Hypertension Caused by Qing-Dai (Chinese Herbal Drug) for Ulcerative Colitis.". Int Heart J 2023; 64:1166. [PMID: 37967984 DOI: 10.1536/ihj.23-281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
|
7
|
Han YH, Kee JY. Extract of Isatidis Radix Inhibits Lipid Accumulation in In Vitro and In Vivo by Regulating Oxidative Stress. Antioxidants (Basel) 2023; 12:1426. [PMID: 37507964 PMCID: PMC10376543 DOI: 10.3390/antiox12071426] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/07/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Isatidis Radix (IR), the root of Isatis tinctoria L. belonging to Brassicaceae, has been traditionally used as a fever reducer. Although some pharmacological effects, such as anti-diabetes, anti-virus, and anti-inflammatory, have been reported, there is no study on the anti-obesity effect of IR. This study used 3T3-L1 cells, human mesenchymal adipose stem cells (hAMSCs), and a high-fat diet (HFD)-induced obese mouse model to confirm the anti-adipogenic effect of IR. Intracellular lipid accumulation in 3T3-L1 cells and hAMSCs was decreased by IR treatment.IR extract especially suppressed reactive oxygen species (ROS) production through a cluster of differentiation 36 (CD36)-AMP-activated protein kinase (AMPK) pathway. Consequently, the expressions of peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT-enhancer-binding proteins alpha (C/EBPα), and fatty acid synthesis (FAS) were inhibited by IR extract. In addition, β-oxidation-related genes were also decreased by treatment of IR extract. IR inhibited weight gain through this cascade in the HFD-induced obese mouse model. IR significantly suppressed lipid accumulation in epididymal white adipose tissue (eWAT). Furthermore, the administration of IR extract decreased serum free fatty acid (FFA), total cholesterol (TC), and LDL cholesterol, suggesting that it could be a potential drug for obesity by inhibiting lipid accumulation.
Collapse
Affiliation(s)
- Yo-Han Han
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ji-Ye Kee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan 54538, Republic of Korea
| |
Collapse
|
8
|
Liu J, Mallick S, Xie Y, Grassin C, Lucas B, Schölermann B, Pahl A, Scheel R, Strohmann C, Protzel C, Berg T, Merten C, Ziegler S, Waldmann H. Morphological Profiling Identifies the Motor Protein Eg5 as Cellular Target of Spirooxindoles. Angew Chem Int Ed Engl 2023; 62:e202301955. [PMID: 36929571 DOI: 10.1002/anie.202301955] [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: 02/08/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/18/2023]
Abstract
Oxindoles and iso-oxindoles are natural product-derived scaffolds that provide inspiration for the design and synthesis of novel biologically relevant compound classes. Notably, the spirocyclic connection of oxindoles with iso-oxindoles has not been explored by nature but promises to provide structurally related compounds endowed with novel bioactivity. Therefore, methods for their efficient synthesis and the conclusive discovery of their cellular targets are highly desirable. We describe a selective RhIII -catalyzed scaffold-divergent synthesis of spirooxindole-isooxindoles and spirooxindole-oxindoles from differently protected diazooxindoles and N-pivaloyloxy aryl amides which includes a functional group-controlled Lossen rearrangement as key step. Unbiased morphological profiling of a corresponding compound collection in the Cell Painting assay efficiently identified the mitotic kinesin Eg5 as the cellular target of the spirooxindoles, defining a unique Eg5 inhibitor chemotype.
Collapse
Affiliation(s)
- Jie Liu
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Street 11, 44227, Dortmund, Germany
| | - Shubhadip Mallick
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Street 11, 44227, Dortmund, Germany
| | - Yusheng Xie
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Street 11, 44227, Dortmund, Germany
| | - Corentin Grassin
- Ruhr University Bochum, Faculty of Chemistry and Biochemistry, Organic Chemistry II, University-Street 150, 44801, Bochum, Germany
| | - Belén Lucas
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Street 11, 44227, Dortmund, Germany
| | - Beate Schölermann
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Street 11, 44227, Dortmund, Germany
| | - Axel Pahl
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Street 11, 44227, Dortmund, Germany
- Compound Management and Screening Center, Otto-Hahn-Street 11, 44227, Dortmund, Germany
| | - Rebecca Scheel
- Technical University Dortmund, Faculty of Chemistry, Inorganic Chemistry, Otto-Hahn-Street 6, 44221, Dortmund, Germany
| | - Carsten Strohmann
- Technical University Dortmund, Faculty of Chemistry, Inorganic Chemistry, Otto-Hahn-Street 6, 44221, Dortmund, Germany
| | - Christoph Protzel
- Leipzig University, Institute of Organic Chemistry, Johannisallee 29, 04103, Leipzig, Germany
| | - Thorsten Berg
- Leipzig University, Institute of Organic Chemistry, Johannisallee 29, 04103, Leipzig, Germany
| | - Christian Merten
- Ruhr University Bochum, Faculty of Chemistry and Biochemistry, Organic Chemistry II, University-Street 150, 44801, Bochum, Germany
| | - Slava Ziegler
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Street 11, 44227, Dortmund, Germany
| | - Herbert Waldmann
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Street 11, 44227, Dortmund, Germany
- Technical University Dortmund, Faculty of Chemistry, Chemical Biology, Otto-Hahn-Street 6, 44221, Dortmund, Germany
| |
Collapse
|
9
|
Miao YB, Zhao W, Renchi G, Gong Y, Shi Y. Customizing delivery nano-vehicles for precise brain tumor therapy. J Nanobiotechnology 2023; 21:32. [PMID: 36707835 PMCID: PMC9883977 DOI: 10.1186/s12951-023-01775-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 01/09/2023] [Indexed: 01/29/2023] Open
Abstract
Although some tumor has become a curable disease for many patients, involvement of the central nervous system (CNS) is still a major concern. The blood-brain barrier (BBB), a special structure in the CNS, protects the brain from bloodborne pathogens via its excellent barrier properties and hinders new drug development for brain tumor. Recent breakthroughs in nanotechnology have resulted in various nanovehicless (NPs) as drug carriers to cross the BBB by different strategys. Here, the complex compositions and special characteristics of causes of brain tumor formation and BBB are elucidated exhaustively. Additionally, versatile drug nanovehicles with their recent applications and their pathways on different drug delivery strategies to overcome the BBB obstacle for anti-brain tumor are briefly discussed. Customizing nanoparticles for brain tumor treatments is proposed to improve the efficacy of brain tumor treatments via drug delivery from the gut to the brain. This review provides a broad perspective on customizing delivery nano-vehicles characteristics facilitate drug distribution across the brain and pave the way for the creation of innovative nanotechnology-based nanomaterials for brain tumor treatments.
Collapse
Affiliation(s)
- Yang-Bao Miao
- grid.410646.10000 0004 1808 0950Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000 China ,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 Sichuan China
| | - Wang Zhao
- grid.410646.10000 0004 1808 0950Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000 China ,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 Sichuan China
| | - Gao Renchi
- grid.410646.10000 0004 1808 0950Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000 China ,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 Sichuan China
| | - Ying Gong
- grid.263901.f0000 0004 1791 7667School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031 People’s Republic of China
| | - Yi Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 Sichuan China ,grid.9227.e0000000119573309Natural Products Research Center, Institute of Chengdu Biology, Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, 610072 Sichuan China ,grid.410646.10000 0004 1808 0950Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, 610072 Sichuan China
| |
Collapse
|
10
|
Combining the In Silico and In Vitro Assays to Identify Strobilanthes cusia Kuntze Bioactives against Penicillin-Resistant Streptococcus pneumoniae. Pharmaceuticals (Basel) 2023; 16:ph16010105. [PMID: 36678602 PMCID: PMC9863409 DOI: 10.3390/ph16010105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
Leaves of Strobilanthes cusia Kuntze (S. cusia) are a widely used alexipharmic Traditional Chinese Medicine (TCM) in southern China for the prevention of cold and respiratory tract infectious diseases. One of the most common bacterial pathogens in the respiratory tract is the gram-positive bacterium Streptococcus pneumoniae. The antibiotic resistance of colonized S. pneumoniae makes it a more serious threat to public health. In this study, the leaves of S. cusia were found to perform antibacterial effects on the penicillin-resistant S. pneumoniae (PRSP). Confocal assay and Transmission Electron Microscopy (TEM) monitored the diminished cell wall integrity and capsule thickness of the PRSP with treatment. The following comparative proteomics analysis revealed that the glycometabolism-related pathways were enriched for the differentially expressed proteins between the samples with treatment and the control. To further delve into the specific single effective compound, the bio-active contents of leaves of S. cusia were analyzed by UPLC-UV-ESI-Q-TOF/MS, and 23 compounds were isolated for anti-PRSP screening. Among them, Tryptanthrin demonstrated the most promising effect, and it possibly inhibited the N-glycan degradation proteins, as suggested by reverse docking analysis in silico and further experimental verification by the surface plasmon resonance assay (SPR). Our study provided a research foundation for applications of the leaves of S. cusia as a TCM, and supplied a bio-active compound Tryptanthrin as a candidate drug skeleton for infectious diseases caused by the PRSP.
Collapse
|
11
|
Zheng Y, Li X, Kuang L, Wang Y. New insights into the characteristics of DRAK2 and its role in apoptosis: From molecular mechanisms to clinically applied potential. Front Pharmacol 2022; 13:1014508. [PMID: 36386181 PMCID: PMC9649744 DOI: 10.3389/fphar.2022.1014508] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/12/2022] [Indexed: 11/27/2022] Open
Abstract
As a member of the death-associated protein kinase (DAPK) family, DAP kinase-associated apoptosis-inducing kinase 2 (DRAK2) performs apoptosis-related functions. Compelling evidence suggests that DRAK2 is involved in regulating the activation of T lymphocytes as well as pancreatic β-cell apoptosis in type I diabetes. In addition, DRAK2 has been shown to be involved in the development of related tumor and non-tumor diseases through a variety of mechanisms, including exacerbation of alcoholic fatty liver disease (NAFLD) through SRSF6-associated RNA selective splicing mechanism, regulation of chronic lymphocytic leukemia and acute myeloid leukemia, and progression of colorectal cancer. This review focuses on the structure, function, and upstream pathways of DRAK2 and discusses the potential and challenges associated with the clinical application of DRAK2-based small-molecule inhibitors, with the aim of advancing DRAK2 research.
Collapse
Affiliation(s)
| | | | | | - Yong Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Traditional Chinese Medicine Regulates Th17/Treg Balance in Treating Inflammatory Bowel Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6275136. [PMID: 36159571 PMCID: PMC9499767 DOI: 10.1155/2022/6275136] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/20/2022] [Indexed: 01/30/2023]
Abstract
Inflammatory bowel disease (IBD), also known as chronic nonspecific inflammatory disease of the colon and rectum, is primarily characterized by mucopurulent bloody stools, diarrhea, abdominal pain, and tenesmus. Its cause is uncertain. IBD patients frequently experience a high rate of recurrence, a protracted treatment course, and a high risk of carcinogenesis. Additionally, the difficulty of treatment is significantly increased by these illness characteristics. Currently, the normal treatment for this illness can lessen symptoms to some amount and even meet clinical treatment requirements, but due to serious side effects, unfavorable reactions, and high costs, we need to develop better complementary and alternative medicines. A number of studies have found that the imbalance of T helper cell 17 (Th17)/regulatory T cells (Treg) contributes significantly to the occurrence and progression of IBD and that Th17/Treg balance restoration is frequently useful in the management of IBD. As a result, regulating the Th17/Treg balance has also emerged as a novel approach to treating IBD. Traditional Chinese medicine (TCM) has gained popularity in recent years due to its advantages of low side effects, a variety of targets, and multiple regulatory mechanisms. A number of studies have shown that TCM can successfully intervene in the Th17/Treg imbalance and restore it, and research on the prevention and treatment of IBD by TCM by restoring Th17/Treg has also shown promising results. The characteristics of the Th17/Treg balance and its role in the pathogenesis of IBD, as well as the role of TCM in regulating the Th17/Treg imbalance, are analyzed. The research results are expected to provide a theoretical basis for the clinical treatment and pathology mechanism research of IBD.
Collapse
|
13
|
Shi J, Yang Y, Zhou X, Zhao L, Li X, Yusuf A, Hosseini MSMZ, Sefidkon F, Hu X. The current status of old traditional medicine introduced from Persia to China. Front Pharmacol 2022; 13:953352. [PMID: 36188609 PMCID: PMC9515588 DOI: 10.3389/fphar.2022.953352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Traditional Chinese medicine (TCM) includes over ten thousand herbal medicines, some of which were introduced from outside countries and territories. The Silk Road enabled the exchange of merchandise such as teas, silks, carpets, and medicines between the East and West of the Eurasia continent. During this time, the ‘Compendium of Materia Medica’ (CMM) was composed by a traditional medicine practitioner, Shizhen Li (1,518–1,593) of the Ming Dynasty. This epoch-making masterpiece collected knowledge of traditional medical materials and treatments in China from the 16th century and before in utmost detail, including the origin where a material was obtained. Of 1892 medical materials from the CMM, 46 came from Persia (now Iran). In this study, the basic information of these 46 materials, including the time of introduction, the medicinal value in TCM theory, together with the current status of these medicines in China and Iran, are summarized. It is found that 20 herbs and four stones out of the 46 materials are registered as medicinal materials in the latest China Pharmacopoeia. Now most of these herbs and stones are distributed in China or replacements are available but saffron, ferula, myrrh, and olibanum are still highly dependent on imports. This study may contribute to the further development, exchange, and internationalization of traditional medicine of various backgrounds in the world, given the barriers of transportation and language are largely eased in nowadays.
Collapse
Affiliation(s)
- Jinmin Shi
- College of Plant Science and Technology, Innovation Academy of International Traditional Chinese Medicinal Materials, National-Regional Joint Engineering Research Center in Hubei for Medicinal Plant Breeding and Cultivation, Medicinal Plant Engineering Research Center of Hubei Province, Institute for Medicinal Plants, Huazhong Agricultural University, Wuhan, China
- Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yifan Yang
- College of Plant Science and Technology, Innovation Academy of International Traditional Chinese Medicinal Materials, National-Regional Joint Engineering Research Center in Hubei for Medicinal Plant Breeding and Cultivation, Medicinal Plant Engineering Research Center of Hubei Province, Institute for Medicinal Plants, Huazhong Agricultural University, Wuhan, China
| | - Xinxin Zhou
- College of Plant Science and Technology, Innovation Academy of International Traditional Chinese Medicinal Materials, National-Regional Joint Engineering Research Center in Hubei for Medicinal Plant Breeding and Cultivation, Medicinal Plant Engineering Research Center of Hubei Province, Institute for Medicinal Plants, Huazhong Agricultural University, Wuhan, China
| | - Lijun Zhao
- Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiaohua Li
- College of Plant Science and Technology, Innovation Academy of International Traditional Chinese Medicinal Materials, National-Regional Joint Engineering Research Center in Hubei for Medicinal Plant Breeding and Cultivation, Medicinal Plant Engineering Research Center of Hubei Province, Institute for Medicinal Plants, Huazhong Agricultural University, Wuhan, China
| | - Abdullah Yusuf
- College of Chemistry and Environmental Science, Laboratory of Xinjiang Native Medicinal and Edible Plant Resources Chemistry. Kashi University, Kashgar, China
| | - Mohaddeseh S. M. Z. Hosseini
- College of Plant Science and Technology, Innovation Academy of International Traditional Chinese Medicinal Materials, National-Regional Joint Engineering Research Center in Hubei for Medicinal Plant Breeding and Cultivation, Medicinal Plant Engineering Research Center of Hubei Province, Institute for Medicinal Plants, Huazhong Agricultural University, Wuhan, China
| | | | - Xuebo Hu
- College of Plant Science and Technology, Innovation Academy of International Traditional Chinese Medicinal Materials, National-Regional Joint Engineering Research Center in Hubei for Medicinal Plant Breeding and Cultivation, Medicinal Plant Engineering Research Center of Hubei Province, Institute for Medicinal Plants, Huazhong Agricultural University, Wuhan, China
- *Correspondence: Xuebo Hu,
| |
Collapse
|
14
|
Xu S, Jin T, Weng J. Endothelial Cells as a Key Cell Type for Innate Immunity: A Focused Review on RIG-I Signaling Pathway. Front Immunol 2022; 13:951614. [PMID: 35865527 PMCID: PMC9294349 DOI: 10.3389/fimmu.2022.951614] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/06/2022] [Indexed: 12/25/2022] Open
Abstract
The vascular endothelium consists of a highly heterogeneous monolayer of endothelial cells (ECs) which are the primary target for bacterial and viral infections due to EC’s constant and close contact with the bloodstream. Emerging evidence has shown that ECs are a key cell type for innate immunity. Like macrophages, ECs serve as sentinels when sensing invading pathogens or microbial infection caused by viruses and bacteria. It remains elusive how ECs senses danger signals, transduce the signal and fulfil immune functions. Retinoic acid-inducible gene-I (RIG-I, gene name also known as DDX58) is an important member of RIG-I-like receptor (RLR) family that functions as an important pathogen recognition receptor (PRR) to execute immune surveillance and confer host antiviral response. Recent studies have demonstrated that virus infection, dsRNA, dsDNA, interferons, LPS, and 25-hydroxycholesterol (25-HC) can increase RIG-1 expression in ECs and propagate anti-viral response. Of translational significance, RIG-I activation can be inhibited by Panax notoginseng saponins, endogenous PPARγ ligand 15-PGJ2, tryptanthrin and 2-animopurine. Considering the pivotal role of inflammation and innate immunity in regulating endothelial dysfunction and atherosclerosis, here we provided a concise review of the role of RIG-I in endothelial cell function and highlight future direction to elucidate the potential role of RIG-I in regulating cardiovascular diseases as well as virus infectious disease, including COVID-19. Furthered understanding of RIG-I-mediated signaling pathways is important to control disorders associated with altered immunity and inflammation in ECs.
Collapse
Affiliation(s)
- Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
- Laboratory of Metabolics and Cardiovascular Diseases, Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province , University of Science and Technology of China, Hefei, China
- *Correspondence: Suowen Xu, ; Jianping Weng,
| | - Tengchuan Jin
- Laboratory of Structural Immunology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jianping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
- Laboratory of Metabolics and Cardiovascular Diseases, Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province , University of Science and Technology of China, Hefei, China
- *Correspondence: Suowen Xu, ; Jianping Weng,
| |
Collapse
|
15
|
Reiss RA, Makhnin O, Lowe TC. Rapid Method to Quantify the Antiviral Potential of Porous and Nonporous Material Using the Enveloped Bacteriophage Phi6. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:8350-8362. [PMID: 35543429 DOI: 10.1021/acs.est.1c07716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The pandemic revealed significant gaps in our understanding of the antiviral potential of porous textiles used for personal protective equipment and nonporous touch surfaces. What is the fate of a microbe when it encounters an abiotic surface? How can we change the microenvironment of materials to improve antimicrobial properties? Filling these gaps requires increasing data generation throughput. A method to accomplish this leverages the use of the enveloped bacteriophage ϕ6, an adjustable spacing multichannel pipette, and the statistical design opportunities inherent in the ordered array of the 24-well culture plate format, resulting in a semi-automated small drop assay. For 100 mm2 nonporous coupons of Cu and Zn, the reduction in ϕ6 infectivity fits first-order kinetics, resulting in half-lives (T50) of 4.2 ± 0.1 and 29.4 ± 1.6 min, respectively. In contrast, exposure to stainless steel has no significant effect on infectivity. For porous textiles, differences associated with composition, color, and surface treatment of samples are detected within 5 min of exposure. Half-lives for differently dyed Zn-containing fabrics from commercially available masks ranged from 2.1 ± 0.05 to 9.4 ± 0.2 min. A path toward full automation and the application of machine learning techniques to guide combinatorial material engineering is presented.
Collapse
Affiliation(s)
- Rebecca A Reiss
- Biology Department, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, New Mexico 87801-4750, United States
| | - Oleg Makhnin
- Mathematics Department, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, New Mexico 87801-4750, United States
| | - Terry C Lowe
- Department of Metallurgical & Materials Engineering, Colorado School of Mines, 920 15th, Street, Golden, Colorado 80401-1887, United States
| |
Collapse
|
16
|
Synthesis of new substituted 7-azaisoindigos. RESULTS IN CHEMISTRY 2022. [DOI: 10.1016/j.rechem.2022.100363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
17
|
Shou X, Wang Y, Zhang X, Zhang Y, Yang Y, Duan C, Yang Y, Jia Q, Yuan G, Shi J, Shi S, Cui H, Hu Y. Network Pharmacology and Molecular Docking Analysis on Molecular Mechanism of Qingzi Zhitong Decoction in the Treatment of Ulcerative Colitis. Front Pharmacol 2022; 13:727608. [PMID: 35237152 PMCID: PMC8883437 DOI: 10.3389/fphar.2022.727608] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
Ulcerative colitis (UC) is a disease with complex pathological mechanisms. We explored the potential molecular mechanisms behind the therapeutic functions of Qingzi Zhitong decoction (QZZTD) in the treatment of UC by network pharmacology and molecular docking. QZZTD is a formula of Chinese traditional medicine consisting of 10 herbs. The potential active ingredients of QZZTD and their target genes were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform database, and UC-related target genes were obtained from GeneCards and OMIM databases. A total of 138 co-identified target genes were obtained by plotting the intersection target Venn diagram, and then the STRING database and Cytoscape software were used to establish protein-protein interaction networks and herb-ingredient-target networks. Four key active compounds and nine key proteins were identified. Then, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses showed that the biological functions of potential target genes were associated with DNA transcription, signaling receptor and ligand activity, cytokine activity, cellular autophagy, and antioxidant pathways, with related pathways involving the phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway, advanced glycosylation end product (AGE)-RAGE signaling pathway, tumor necrosis factor (TNF) signaling pathway, and IL-17 signaling pathway. Moreover, the binding activities of key target genes and essential active compounds of Chinese herbal medicines in QZZTD were further validated by molecular docking. This demonstrated that quercetin, luteolin, hyndarin, and beta-sitosterol had good binding to eight key proteins, and Akt1 was the target protein with the best binding activity, suggesting that Akt1 could be the essential mediator responsible for signaling transduction after QZZTD administration. The rat experiment verified that QZZTD inhibited PI3K-Akt pathway activation and reduced inflammation in UC. In conclusion, our study suggested four potential key active components, including quercetin, were identified in QZZTD, which could interact with Akt1 and modulate the activation of the PI3K-Akt pathway. The other three pathways may also be involved in the signaling transduction induced by QZZTD in the treatment of UC.
Collapse
Affiliation(s)
- Xintian Shou
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Yumeng Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Xuesong Zhang
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Yanju Zhang
- National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yan Yang
- National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Chenglin Duan
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Yihan Yang
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Qiulei Jia
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Guozhen Yuan
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Jingjing Shi
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Shuqing Shi
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Hanming Cui
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Yuanhui Hu
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| |
Collapse
|
18
|
Lee CL, Wang CM, Yen HR, Song YC, Chen CJ. Indigodole E from Strobilanthes cusia exhibits anti-IL-17A effect. Nat Prod Res 2022; 36:5935-5939. [PMID: 35192387 DOI: 10.1080/14786419.2022.2041633] [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] [Indexed: 10/19/2022]
Abstract
One new indazole alkaloid, indigodole E (1), was isolated from a traditional Chinese medicine Qing Dai prepared from the aerial parts of Strobilanthes cusia. The structure of 1 was elucidated by NMR, MS, UV, and IR spectra as well as optical rotation. Additionally, compound 1 could obviously inhibit not only IL-17A protein production at concentrations from 1.25 to 2.5 μg/mL, but also IL-17 gene expression at concentrations from 5.0 to 10.0 μg/mL without cytotoxicity toward Th17 and Jukat cells, respectively. Overall, indazole analogue 1 could be the anti-IL 17 A contributor of Qing Dai in this investigation.
Collapse
Affiliation(s)
- Chia-Lin Lee
- Department of Cosmeceutics, China Medical University, Taichung, Taiwan.,Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Chien-Ming Wang
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Hung-Rong Yen
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan.,Research Center for Traditional Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ying-Chyi Song
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Chao-Jung Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.,Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
19
|
Rahman M, Almalki WH, Panda SK, Das AK, Alghamdi S, Soni K, Hafeez A, Handa M, Beg S, Rahman Z. Therapeutic application of microsponges based drug delivery system. Curr Pharm Des 2022; 28:595-608. [PMID: 35040411 DOI: 10.2174/1381612828666220118121536] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 12/23/2021] [Indexed: 11/22/2022]
Abstract
Microsponges delivery system (MDS) is highly porous, cross-linked based polymeric systems, that activates in the presence of temperature, rubbing and pH. MDS offers wide range of advantage, like controlled drug release, site-specific action, stable over a broad range of pH, poor irritation, cost effective, improved patient compliance. They can be transformed into various dosage forms like creams, gels, and lotions. MDS based system are suitable for treatment of topical disorders like acne, psoriasis, dandruff, eczema, scleroderma, hair loss, skin cancer, and other dreadful diseases. MDS application for drug delivery is not limited to topical drug delivery but also explored for oral, parenteral and pulmonary drug delivery. Microsponges were studied for colon targeting of drugs and genes. Additionally, MDS has wide application for sunscreen, cosmetics, and over the counter (OTC) products. Furthermore, MDS does not actuate any irritation, genotoxicity, immunogenicity or cytotoxicity. Therefore, this review extensively highlights about microsponges, their advantages, key factors affecting the micro-sponges' characteristics, the therapeutic application of microsponges in topical disorders, cancer, as cosmetics, recent advances in MDS and addresses the associated challenges.
Collapse
Affiliation(s)
- Mahfoozur Rahman
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology &Sciences, Allahabad, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Saudi Arabia
| | - Sunil K Panda
- Menovo Pharmaceuticals Research Lab, Ningbo, People\'s Republic of China
| | - Aman Kumar Das
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology &Sciences, Allahabad, India
| | - Saad Alghamdi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Kirti Soni
- Formulation Development, Dabur Research Foundation, 22 Site IV Sahibabad Industrial Area, Ghaziabad, Uttar Pradesh, India
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh, India
| | - Mayank Handa
- Department of Pharmaceutics, NIPER, Raebareli, Lucknow, Uttar Pradesh, 226002
| | - Sarwar Beg
- School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UKb
| | - Ziyaur Rahman
- Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Texas A&M University, College Station , TX 77843
| |
Collapse
|
20
|
Indigo Pulverata Levis (Chung-Dae, Persicaria tinctoria) Alleviates Atopic Dermatitis-like Inflammatory Responses In Vivo and In Vitro. Int J Mol Sci 2022; 23:ijms23010553. [PMID: 35008979 PMCID: PMC8745452 DOI: 10.3390/ijms23010553] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/30/2021] [Accepted: 01/01/2022] [Indexed: 12/31/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease associated with a type 2 T helper cell (Th2) immune response. The IndigoPulverata Levis extract (CHD) is used in traditional Southeast Asian medicine; however, its beneficial effects on AD remain uninvestigated. Therefore, we investigated the therapeutic effects of CHD in 2,4-dinitrochlorobenzene (DNCB)-induced BALB/c mice and tumor necrosis factor (TNF)-α- and interferon gamma (IFN)-γ-stimulated HaCaT cells. We evaluated immune cell infiltration, skin thickness, and the serum IgE and TNF-α levels in DNCB-induced AD mice. Moreover, we measured the expression levels of pro-inflammatory cytokines, mitogen-activated protein kinase (MAPK), and the nuclear factor-kappa B (NF-κB) in the mice dorsal skin. We also studied the effect of CHD on the translocation of NF-κB p65 and inflammatory chemokines in HaCaT cells. Our in vivo results revealed that CHD reduced the dermis and epidermis thicknesses and inhibited immune cell infiltration. Furthermore, it suppressed the proinflammatory cytokine expression and MAPK and NF-κB phosphorylations in the skin tissue and decreased serum IgE and TNF-α levels. In vitro results indicated that CHD downregulated inflammatory chemokines and blocked NF-κB p65 translocation. Thus, we deduced that CHD is a potential drug candidate for AD treatment.
Collapse
|
21
|
Czapka A, Grune C, Schädel P, Bachmann V, Scheuer K, Dirauf M, Weber C, Skaltsounis AL, Jandt KD, Schubert US, Fischer D, Werz O. Drug delivery of 6-bromoindirubin-3'-glycerol-oxime ether employing poly(D,L-lactide-co-glycolide)-based nanoencapsulation techniques with sustainable solvents. J Nanobiotechnology 2022; 20:5. [PMID: 34983538 PMCID: PMC8725458 DOI: 10.1186/s12951-021-01179-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Insufficient solubility and stability of bioactive small molecules as well as poor biocompatibility may cause low bioavailability and are common obstacles in drug development. One example of such problematic molecules is 6-bromoindirubin-3'-glycerol-oxime ether (6BIGOE), a hydrophobic indirubin derivative. 6BIGOE potently modulates the release of inflammatory cytokines and lipid mediators from isolated human monocytes through inhibition of glycogen synthase kinase-3 in a favorable fashion. However, 6BIGOE suffers from poor solubility and short half-lives in biological aqueous environment and exerts cytotoxic effects in various mammalian cells. In order to overcome the poor water solubility, instability and cytotoxicity of 6BIGOE, we applied encapsulation into poly(D,L-lactide-co-glycolide) (PLGA)-based nanoparticles by employing formulation methods using the sustainable solvents Cyrene™ or 400 g/mol poly(ethylene glycol) as suitable technology for efficient drug delivery of 6BIGOE. RESULTS For all preparation techniques the physicochemical characterization of 6BIGOE-loaded nanoparticles revealed comparable crystallinity, sizes of about 230 nm with low polydispersity, negative zeta potentials around - 15 to - 25 mV, and biphasic release profiles over up to 24 h. Nanoparticles with improved cellular uptake and the ability to mask cytotoxic effects of 6BIGOE were obtained as shown in human monocytes over 48 h as well as in a shell-less hen's egg model. Intriguingly, encapsulation into these nanoparticles fully retains the anti-inflammatory properties of 6BIGOE, that is, favorable modulation of the release of inflammation-relevant cytokines and lipid mediators from human monocytes. CONCLUSIONS Our formulation method of PLGA-based nanoparticles by applying sustainable, non-toxic solvents is a feasible nanotechnology that circumvents the poor bioavailability and biocompatibility of the cargo 6BIGOE. This technology yields favorable drug delivery systems for efficient interference with inflammatory processes, with improved pharmacotherapeutic potential.
Collapse
Affiliation(s)
- Anna Czapka
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743, Jena, Germany
| | - Christian Grune
- Pharmaceutical Technology and Biopharmacy, Institute of Pharmacy, Friedrich Schiller University Jena, Lessingstraße 8, 07743, Jena, Germany
| | - Patrick Schädel
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743, Jena, Germany
| | - Vivien Bachmann
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743, Jena, Germany
| | - Karl Scheuer
- Chair of Materials Science (CMS), Faculty of Physics and Astronomy, Otto Schott Institute of Materials Research, Friedrich Schiller University Jena, Löbdergraben 32, 07743, Jena, Germany
| | - Michael Dirauf
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Christine Weber
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Alexios-Leandros Skaltsounis
- Department of Pharmacy, Division of Pharmacognosy and Natural Products Chemistry, University of Athens, Panepistimiopolis Zografou, 15771, Athens, Greece
| | - Klaus D Jandt
- Chair of Materials Science (CMS), Faculty of Physics and Astronomy, Otto Schott Institute of Materials Research, Friedrich Schiller University Jena, Löbdergraben 32, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Ulrich S Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Dagmar Fischer
- Pharmaceutical Technology and Biopharmacy, Institute of Pharmacy, Friedrich Schiller University Jena, Lessingstraße 8, 07743, Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany.
- Division of Pharmaceutical Technology, Department for Chemistry and Pharmacy, Friedrich-Alexander-University Erlangen-Nürnberg, Cauerstrasse 4, 91058, Erlangen, Germany.
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743, Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany.
| |
Collapse
|
22
|
Shi J, Weng JH, Mitchison TJ. Immunomodulatory drug discovery from herbal medicines: Insights from organ-specific activity and xenobiotic defenses. eLife 2021; 10:e73673. [PMID: 34779403 PMCID: PMC8592567 DOI: 10.7554/elife.73673] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Traditional herbal medicines, which emphasize a holistic, patient-centric view of disease treatment, provide an exciting starting point for discovery of new immunomodulatory drugs. Progress on identification of herbal molecules with proven single agent activity has been slow, in part because of insufficient consideration of pharmacology fundamentals. Many molecules derived from medicinal plants exhibit low oral bioavailability and rapid clearance, leading to low systemic exposure. Recent research suggests that such molecules can act locally in the gut or liver to activate xenobiotic defense pathways that trigger beneficial systemic effects on the immune system. We discuss this hypothesis in the context of four plant-derived molecules with immunomodulatory activity: indigo, polysaccharides, colchicine, and ginsenosides. We end by proposing research strategies for identification of novel immunomodulatory drugs from herbal medicine sources that are informed by the possibility of local action in the gut or liver, leading to generation of systemic immune mediators.
Collapse
Affiliation(s)
- Jue Shi
- Centre for Quantitative Systems Biology, Department of Physics and Department of Biology, Hong Kong Baptist UniversityHong KongChina
| | - Jui-Hsia Weng
- Department of Systems Biology, Harvard Medical SchoolBostonUnited States
- Institute of Biological Chemistry, Academia SinicaTaipeiTaiwan
| | | |
Collapse
|