1
|
Radu A, Tit DM, Endres LM, Radu AF, Vesa CM, Bungau SG. Naturally derived bioactive compounds as precision modulators of immune and inflammatory mechanisms in psoriatic conditions. Inflammopharmacology 2025; 33:527-549. [PMID: 39576422 PMCID: PMC11842495 DOI: 10.1007/s10787-024-01602-z] [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: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 02/22/2025]
Abstract
Psoriasis represents a chronic autoimmune skin condition defined by various clinical forms, including inverse, erythrodermic, pustular, guttate, plaque types. While current therapies, including topical treatments but also systemic through conventional synthetic drugs and biologics, have improved symptom management, no treatment completely cures the disease, and numerous options are linked to considerable adverse effects, including immunosuppression and carcinogenic risks. Therefore, there is growing interest in bioactive compounds from natural sources due to their potential to reduce inflammation and oxidative stress in psoriasis with fewer adverse effects. The present narrative review aimed to address the limitations of current psoriasis therapies by exploring the therapeutic potential of bioactive compounds in the classes of flavonoids, terpenoids, omega-3 fatty acids, and alkaloids assessed through complex experimental models, focusing on their immunomodulatory and anti-inflammatory properties. Recent studies highlight the efficacy of natural bioactive compounds in reducing psoriasis symptoms, either as standalone treatments or in combination with conventional therapies. While these compounds show promise in alleviating psoriasis-related inflammation, further research is needed to optimize their therapeutic use, understand their mechanisms of action, and assess long-term safety. Future studies should focus on clinical trials to establish standardized protocols for incorporating bioactive compounds into psoriasis management and explore their potential role in personalized treatment strategies. Continued research is essential to develop more effective, safer, and affordable therapeutic options for psoriasis patients.
Collapse
Affiliation(s)
- Ada Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087, Oradea, Romania
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028, Oradea, Romania
| | - Delia Mirela Tit
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087, Oradea, Romania
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028, Oradea, Romania
| | - Laura Maria Endres
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087, Oradea, Romania
- Department of Psycho-Neurosciences and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410073, Oradea, Romania
| | - Andrei-Flavius Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087, Oradea, Romania.
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073, Oradea, Romania.
| | - Cosmin Mihai Vesa
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087, Oradea, Romania.
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073, Oradea, Romania.
| | - Simona Gabriela Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087, Oradea, Romania
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028, Oradea, Romania
| |
Collapse
|
2
|
Zhang G, Jiang Y, Wang Z, Guo Z, Hu J, Li X, Wang Y, Jing Z. FUS/circZEB1/miR-128-3p/LBH feedback loop contributes to the malignant phenotype of GSCs via TNF-α-mediated NF-κB signaling pathway. Cancer Cell Int 2024; 24:365. [PMID: 39511561 PMCID: PMC11545228 DOI: 10.1186/s12935-024-03526-8] [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: 01/09/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
Glioblastoma (GBM) is the most lethal and common primary tumor of central nervous system with a poor prognosis. Glioma stem cells (GSCs) are particularly significant in GBM proliferation, invasion, self-renewal and recurrence. Circular RNAs (circRNAs) play important roles in various physiological and pathological processes, including regulating the biological behavior of GBM. Therefore, discovering novel circRNAs related to GSCs may contribute to a promising approach for treatment of GBM. Herein, we find out a novel circRNA termed circZEB1 with a high expression in glioma. Limb-bud and heart (LBH) is a transcription cofactor and promotes glioma stem cell tumorigenicity in our study. Mechanistically, circZEB1 can upregulate the expression of transcription cofactor LBH via sponging miR-128-3p in GSCs. LBH can facilitate the expression of tumor necrosis factor-α (TNF-α), thus activating the NF-κB signaling pathway to promote the glioma progression. Meanwhile, LBH can also upregulate the RNA binding protein Fused in Sarcoma (FUS) expression, which can bind to and maintain the stability of circZEB1. A positive feedback loop is formed among FUS, circZEB1, miR-128-3p and LBH in GSCs. Our study uncovers a critical role of circZEB1 and provides a novel biomarker for treating GBM.
Collapse
Affiliation(s)
- Guoqing Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China
| | - Yang Jiang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072, Shanghai, People's Republic of China
| | - Zhichao Wang
- Department of Neurosurgery, The People's Hospital of China Medical University, Shenyang, 110067, China
| | - Zhengting Guo
- Department of Neurosurgery, The First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China
| | - Jinpeng Hu
- Department of Neurosurgery, The First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China
| | - Xinqiao Li
- Department of Neurosurgery, The First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China
| | - Yongfeng Wang
- Department of Radiology, The First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China.
| | - Zhitao Jing
- Department of Neurosurgery, The First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China.
| |
Collapse
|
3
|
do Amaral SR, Amantino CF, Atanasov A, Sousa SO, Moakes R, Oliani SM, Grover LM, Primo FL. Photodynamic Therapy as a Novel Therapeutic Modality Applying Quinizarin-Loaded Nanocapsules and 3D Bioprinting Skin Permeation for Inflammation Treatment. Pharmaceuticals (Basel) 2024; 17:1169. [PMID: 39338332 PMCID: PMC11434822 DOI: 10.3390/ph17091169] [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: 07/30/2024] [Revised: 08/27/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Skin inflammation associated with chronic diseases involves a direct role of keratinocytes in its immunopathogenesis, triggering a cascade of immune responses. Despite this, highly targeted treatments remain elusive, highlighting the need for more specific therapeutic strategies. In this study, nanocapsules containing quinizarin (QZ/NC) were developed and evaluated in an in vitro model of keratinocyte-mediated inflammation, incorporating the action of photodynamic therapy (PDT) and analyzing permeation in a 3D skin model. Comprehensive physicochemical, stability, cytotoxicity, and permeation analyses of the nanomaterials were conducted. The nanocapsules demonstrated desirable physicochemical properties, remained stable throughout the analysis period, and exhibited no spectroscopic alterations. Cytotoxicity tests revealed no toxicity at the lowest concentrations of QZ/NC. Permeation and cellular uptake studies confirmed QZ/NC permeation in 3D skin models, along with intracellular incorporation and internalization of the drug, thereby enhancing its efficacy in drug delivery. The developed model for inducing the inflammatory process in vitro yielded promising results, particularly when the synthesized nanomaterial was combined with PDT, showing a reduction in cytokine levels. These findings suggest a potential new therapeutic approach for treating inflammatory skin diseases.
Collapse
Affiliation(s)
- Stéphanie R. do Amaral
- Department of Bioprocess and Biotechnology Engineering, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (S.R.d.A.); (C.F.A.)
| | - Camila F. Amantino
- Department of Bioprocess and Biotechnology Engineering, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (S.R.d.A.); (C.F.A.)
- São Paulo Federal Institute of Education, Science and Technology (IFSP), Matão 15991-502, SP, Brazil
| | - Aleksandar Atanasov
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK; (A.A.); (R.M.); (L.M.G.)
| | - Stefanie Oliveira Sousa
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), São José do Rio Preto 15054-000, SP, Brazil; (S.O.S.); (S.M.O.)
| | - Richard Moakes
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK; (A.A.); (R.M.); (L.M.G.)
| | - Sonia Maria Oliani
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), São José do Rio Preto 15054-000, SP, Brazil; (S.O.S.); (S.M.O.)
| | - Liam M. Grover
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK; (A.A.); (R.M.); (L.M.G.)
| | - Fernando L. Primo
- Department of Bioprocess and Biotechnology Engineering, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (S.R.d.A.); (C.F.A.)
| |
Collapse
|
4
|
Burlec AF, Hăncianu M, Ivănescu B, Macovei I, Corciovă A. Exploring the Therapeutic Potential of Natural Compounds in Psoriasis and Their Inclusion in Nanotechnological Systems. Antioxidants (Basel) 2024; 13:912. [PMID: 39199158 PMCID: PMC11352172 DOI: 10.3390/antiox13080912] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
Psoriasis is a chronic inflammatory disease that affects around 2-3% of the world's population. The treatment for this autoimmune disease still remains centered around conventional methods using synthetic substances, even though more recent advancements focus on biological therapies. Given the numerous side effects of such treatments, current research involves plant extracts and constituents that could prove useful in treating psoriasis. The aim of this narrative review is to highlight the most known representatives belonging to classes of natural compounds such as polyphenols (e.g., astilbin, curcumin, hesperidin, luteolin, proanthocyanidins, and resveratrol), alkaloids (e.g., berberine, capsaicin, and colchicine), coumarins (psoralen and 8-methoxypsoralen), and terpenoids (e.g., celastrol, centelloids, and ursolic acid), along with plants used in traditional medicine that could present therapeutic potential in psoriasis. The paper also provides an overview of these compounds' mechanisms of action and current inclusion in clinical studies, as well as an investigation into their potential incorporation in various nanotechnological systems, such as lipid-based nanocarriers or polymeric nanomaterials, that may optimize their efficacy during treatment.
Collapse
Affiliation(s)
- Ana Flavia Burlec
- Department of Drug Analysis, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.F.B.); (A.C.)
| | - Monica Hăncianu
- Department of Pharmacognosy, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania;
| | - Bianca Ivănescu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Irina Macovei
- Department of Drug Analysis, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.F.B.); (A.C.)
| | - Andreia Corciovă
- Department of Drug Analysis, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.F.B.); (A.C.)
| |
Collapse
|
5
|
Chen Y, Song Y, Wang Z, Lai Y, Yin W, Cai Q, Han M, Cai Y, Xue Y, Chen Z, Li X, Chen J, Li M, Li H, He R. The chemerin-CMKLR1 axis in keratinocytes impairs innate host defense against cutaneous Staphylococcus aureus infection. Cell Mol Immunol 2024; 21:533-545. [PMID: 38532043 PMCID: PMC11143357 DOI: 10.1038/s41423-024-01152-y] [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: 07/04/2023] [Accepted: 03/01/2024] [Indexed: 03/28/2024] Open
Abstract
The skin is the most common site of Staphylococcus aureus infection, which can lead to various diseases, including invasive and life-threatening infections, through evasion of host defense. However, little is known about the host factors that facilitate the innate immune evasion of S. aureus in the skin. Chemerin, which is abundantly expressed in the skin and can be activated by proteases derived from S. aureus, has both direct bacteria-killing activity and immunomodulatory effects via interactions with its receptor CMKLR1. Here, we demonstrate that a lack of the chemerin/CMKLR1 axis increases the neutrophil-mediated host defense against S. aureus in a mouse model of cutaneous infection, whereas chemerin overexpression, which mimics high levels of chemerin in obese individuals, exacerbates S. aureus cutaneous infection. Mechanistically, we identified keratinocytes that express CMKLR1 as the main target of chemerin to suppress S. aureus-induced IL-33 expression, leading to impaired skin neutrophilia and bacterial clearance. CMKLR1 signaling specifically inhibits IL-33 expression induced by cell wall components but not secreted proteins of S. aureus by inhibiting Akt activation in mouse keratinocytes. Thus, our study revealed that the immunomodulatory effect of the chemerin/CMKLR1 axis mediates innate immune evasion of S. aureus in vivo and likely increases susceptibility to S. aureus infection in obese individuals.
Collapse
Affiliation(s)
- Yu Chen
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yan Song
- Department of Laboratory Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Zhe Wang
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Yangfan Lai
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Wei Yin
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Qian Cai
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Miaomiao Han
- Allergy Center, Department of Otolaryngology, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yiheng Cai
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Yushan Xue
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Zhengrong Chen
- Department of Respiratory Diseases, Children's Hospital of Soochow University, Suzhou, China
| | - Xi Li
- Biology Science Institutes, Chongqing Medical University, Chongqing, 400032, China
| | - Jing Chen
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of Nephrology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Min Li
- Department of Laboratory Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Faculty of Medical Laboratory Science, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Huabin Li
- Allergy Center, Department of Otolaryngology, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, 200031, China.
| | - Rui He
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Research Center of Allergy and Diseases, Fudan University, 200040, Shanghai, China.
| |
Collapse
|
6
|
Zhu ZB, Liu MJ, Wang J, Shu Z, Cao J. Secoemestrin C Ameliorates Psoriasis-like Skin Inflammation in Mice by Suppressing the TNF-α/NF-κB Signaling Pathway. Curr Med Sci 2024; 44:232-240. [PMID: 38393530 DOI: 10.1007/s11596-024-2828-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 12/03/2023] [Indexed: 02/25/2024]
Abstract
OBJECTIVE Secoemestrin C (SC), an epitetrathiodioxopiperazine isolated from Aspergillus nidulans, has been previously reported to have immunomodulatory and hepatoprotective effects against acute autoimmune hepatitis. However, the effect of SC on regulating the inflammation and its underlying mechanisms in the pathogenesis of psoriasis remain unclear. This study aimed to evaluate the effects of SC on inflammatory dermatosis both in vitro and in vivo. METHODS In vitro, HaCaT cells were induced with tumor necrosis factor-alpha (TNF-α, 10 ng/mL) to establish an inflammatory injury model, and the expression of nuclear transcription factor-κB (NF-κB) pathway components was measured using qRT-PCR and Western blotting. An in vivo mouse model of imiquimod (IMQ)-induced psoriasis-like skin inflammation was used to evaluate the effectiveness of SC in alleviating psoriasis. RESULTS SC significantly blocked the activation of NF-κB signaling in TNF-α-stimulated HaCaT cells. In addition, systemic and local administration of SC improved psoriatic dermatitis in the IMQ-induced mouse model. SC reduced skin scale and significantly inhibited the secretion of inflammatory factors in skin lesions. CONCLUSION The protective effect of SC against psoriatic-associated inflammation reveals its potential therapeutic value for treating psoriasis.
Collapse
Affiliation(s)
- Zhi-Bin Zhu
- Department of Stomatology, Chengdu Seventh People's Hospital, Chengdu, 610044, China
| | - Meng-Jie Liu
- Department of Stomatology, Chengdu Seventh People's Hospital, Chengdu, 610044, China
| | - Jing Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhou Shu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jie Cao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
7
|
He K, Wang Z, Liu M, Du W, Yin T, Bai R, Duan Q, Wang Y, Lei H, Zheng Y. Exploring the Effect of Xiao-Chai-Hu Decoction on Treating Psoriasis Based on Network Pharmacology and Experiment Validation. Curr Pharm Des 2024; 30:215-229. [PMID: 38532341 DOI: 10.2174/0113816128288527240108110844] [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: 10/20/2023] [Accepted: 12/27/2023] [Indexed: 03/28/2024]
Abstract
BACKGROUND Psoriasis is a chronic, inflammatory and recurrent skin disease. Xiao-Chai-Hu Decoction (XCHD) has shown good effects against some inflammatory diseases and cancers. However, the pharmacological effect and mechanisms of XCHD on psoriasis are not yet clear. OBJECTIVE To uncover the effect and mechanisms of XCHD on psoriasis by integrating network pharmacology, molecular docking, and in vivo experiments. METHODS The active ingredients and corresponding targets of XCHD were screened through Traditional Chinese Medicine Systems Pharmacology Database and Analysis (TCMSP) and Traditional Chinese Medicine Integrated Database (TCMID). Differentially expressed genes (DEGs) of psoriasis were obtained from the gene expression omnibus (GEO) database. The XCHD-psoriasis intersection targets were obtained by intersecting XCHD targets, and DEGs were used to establish the "herb-active ingredient-target" network and Protein-Protein Interaction (PPI) Network. The hub targets were identified based on the PPI network by Cytoscape software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed next. Molecular docking was executed via AutoDockTools-1.5.6. Finally, in vivo experiments were carried out further to validate the therapeutic effects of XCHD on psoriasis. RESULTS 58 active components and 219 targets of XCHD were screened. 4 top-active components (quercetin, baicalein, wogonin and kaempferol) and 7 hub targets (IL1B, CXCL8, CCND1, FOS, MMP9, STAT1 and CCL2) were identified. GO and KEGG pathway enrichment analyses indicated that the TNF signaling pathway, IL-17 signaling pathway and several pathways were involved. Molecular docking results indicated that hub genes had a good affinity to the corresponding key compounds. In imiquimod (IMQ)-induced psoriasis mouse models, XCHD could significantly improve psoriasis-like skin lesions, downregulate KRT17 and Ki67, and inhibit inflammation cytokines and VEGF. CONCLUSION XCHD showed the therapeutic effect on psoriasis by regulating keratinocyte differentiation, and suppressing inflammation and angiogenesis, which provided a theoretical basis for further experiments and clinical research.
Collapse
Affiliation(s)
- Ke He
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Ziyang Wang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Meng Liu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Wenqian Du
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Tingyi Yin
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Ruimin Bai
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Qiqi Duan
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuqian Wang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Hao Lei
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yan Zheng
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
8
|
Zhong Y, Zhang BW, Li JT, Zeng X, Pei JX, Zhang YM, Yang YX, Li FL, Deng Y, Zhao Q. Ethanol extract of Herpetospermum caudigerum Wall ameliorates psoriasis-like skin inflammation and promotes degradation of keratinocyte-derived ICAM-1 and CXCL9. JOURNAL OF INTEGRATIVE MEDICINE 2023; 21:584-592. [PMID: 37989697 DOI: 10.1016/j.joim.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/25/2023] [Indexed: 11/23/2023]
Abstract
OBJECTIVE To explore whether the ethanol extract of Herpetospermum caudigerum Wall (EHC), a Xizang medicinal plant traditionally used for treating liver diseases, can improve imiquimod-induced psoriasis-like skin inflammation. METHODS Immunohistochemistry and immunofluorescence staining were used to determine the effects of topical EHC use in vivo on the skin pathology of imiquimod-induced psoriasis in mice. The protein levels of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and interleukin-17A (IL-17A) in mouse skin samples were examined using immunohistochemical staining. In vitro, IFN-γ-induced HaCaT cells with or without EHC treatment were used to evaluate the expression of keratinocyte-derived intercellular cell adhesion molecule-1 (ICAM-1) and chemokine CXC ligand 9 (CXCL9) using Western blotting and reverse transcription-quantitative polymerase chain reaction. The protein synthesis inhibitor cycloheximide and proteasome inhibitor MG132 were utilized to validate the EHC-mediated mechanism underlying degradation of ICAM-1 and CXCL9. RESULTS EHC improved inflammation in the imiquimod-induced psoriasis mouse model and reduced the levels of IFN-γ, TNF-α, and IL-17A in psoriatic lesions. Treatment with EHC also suppressed ICAM-1 and CXCL9 in epidermal keratinocytes. Further mechanistic studies revealed that EHC suppressed keratinocyte-derived ICAM-1 and CXCL9 by promoting ubiquitin-proteasome-mediated protein degradation rather than transcriptional repression. Seven primary compounds including ehletianol C, dehydrodiconiferyl alcohol, herpetrione, herpetin, herpetotriol, herpetetrone and herpetetrol were identified from the EHC using ultra-performance liquid chromatography-quadrupole-time of flight-mass spectrometry. CONCLUSION Topical application of EHC ameliorates psoriasis-like skin symptoms and improves the inflammation at the lesion sites. Please cite this article as: Zhong Y, Zhang BW, Li JT, Zeng X, Pei JX, Zhang YM, Yang YX, Li FL, Deng Y, Zhao Q. Ethanol extract of Herpetospermum caudigerum Wall ameliorates psoriasis-like skin inflammation and promotes degradation of keratinocyte-derived ICAM-1 and CXCL9. J Integr Med. 2023; 21(6): 584-592.
Collapse
Affiliation(s)
- Ya Zhong
- Engineering Research Center of Sichuan-Xizang Traditional Medicinal Plant, Chengdu University, Chengdu 610106, Sichuan Province, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, Sichuan Province, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, Sichuan Province, China
| | - Bo-Wen Zhang
- Engineering Research Center of Sichuan-Xizang Traditional Medicinal Plant, Chengdu University, Chengdu 610106, Sichuan Province, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, Sichuan Province, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, Sichuan Province, China
| | - Jin-Tao Li
- Engineering Research Center of Sichuan-Xizang Traditional Medicinal Plant, Chengdu University, Chengdu 610106, Sichuan Province, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, Sichuan Province, China; School of Pharmacy, Chengdu University, Chengdu 610106, Sichuan Province, China
| | - Xin Zeng
- Engineering Research Center of Sichuan-Xizang Traditional Medicinal Plant, Chengdu University, Chengdu 610106, Sichuan Province, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, Sichuan Province, China; School of Pharmacy, Chengdu University, Chengdu 610106, Sichuan Province, China
| | - Jun-Xia Pei
- Engineering Research Center of Sichuan-Xizang Traditional Medicinal Plant, Chengdu University, Chengdu 610106, Sichuan Province, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, Sichuan Province, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, Sichuan Province, China
| | - Ya-Mei Zhang
- Key Laboratory of Clinical Genetics, Affiliated Hospital of Chengdu University, Chengdu 610106, Sichuan Province, China
| | - Yi-Xi Yang
- Engineering Research Center of Sichuan-Xizang Traditional Medicinal Plant, Chengdu University, Chengdu 610106, Sichuan Province, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, Sichuan Province, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, Sichuan Province, China
| | - Fu-Lun Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yu Deng
- Engineering Research Center of Sichuan-Xizang Traditional Medicinal Plant, Chengdu University, Chengdu 610106, Sichuan Province, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, Sichuan Province, China; School of Basic Medical Sciences, Chengdu University, Chengdu 610106, Sichuan Province, China.
| | - Qi Zhao
- Engineering Research Center of Sichuan-Xizang Traditional Medicinal Plant, Chengdu University, Chengdu 610106, Sichuan Province, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, Sichuan Province, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, Sichuan Province, China.
| |
Collapse
|
9
|
Wang M, Gao M, Yi Z. Biological effects of IL-33/ST2 axis on oral diseases: autoimmune diseases and periodontal diseases. Int Immunopharmacol 2023; 122:110524. [PMID: 37393839 DOI: 10.1016/j.intimp.2023.110524] [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: 05/09/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 07/04/2023]
Abstract
IL-33 is a relatively new member of the IL-1 cytokine family, which plays a unique role in autoimmune diseases, particularly some oral diseases dominated by immune factors. The IL-33/ST2 axis is the main pathway by which IL-33 signals affect downstream cells to produce an inflammatory response or tissue repair. As a newly discovered pro-inflammatory cytokine, IL-33 can participate in the pathogenesis of autoimmune oral diseases such as Sjogren's syndrome and Behcet's disease. Moreover, the IL-33/ST2 axis also recruits and activates mast cells in periodontitis, producing inflammatory chemokines and mediating gingival inflammation and alveolar bone destruction. Interestingly, the high expression of IL-33 in the alveolar bone, which exhibits anti-osteoclast effects under appropriate mechanical loading, also confirms its dual role of destruction and repair in an immune-mediated periodontal environment. This study reviewed the biological effects of IL-33 in autoimmune oral diseases, periodontitis and periodontal bone metabolism, and elaborated its potential role and impact as a disease enhancer or a repair factor.
Collapse
Affiliation(s)
- Mingfeng Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Mingcen Gao
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Zhe Yi
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China.
| |
Collapse
|
10
|
Peng M, Li J, Zhou J, Zhang B, Liao J, Yang D, Wang Y, Yang Y, Li R, Tang X, Lu Q, Zhao Q. Total alkaloids of Fritillaria unibracteata var. wabuensis bulbus ameliorate chronic asthma via the TRPV1/Ca 2+/NFAT pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154946. [PMID: 37421766 DOI: 10.1016/j.phymed.2023.154946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/07/2023] [Accepted: 06/27/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND Asthma is a chronic inflammatory disease that is challenging to treat. Fritillaria unibracteata var. wabuensis (FUW) is the plant origin for the famous Chinese antitussive medicine Fritillaria Cirrhosae Bulbus. The total alkaloids of Fritillaria unibracteata var. wabuensis bulbus (TAs-FUW) have anti-inflammatory properties and may be used to treat asthma. PURPOSE To explore whether TAs-FUW have bioactivity against airway inflammation and a therapeutic effect on chronic asthma. METHODS The alkaloids were extracted via ultrasonication in a cryogenic chloroform-methanol solution after ammonium-hydroxide percolation of the bulbus. UPLC-Q-TOF/MS was used to characterize the composition of TAs-FUW. An ovalbumin (OVA)-induced asthmatic mouse model was established. We used whole-body plethysmography, ELISA, western blotting, RT-qPCR, and histological analyses to assess the pulmonary pathological changes in these mice after TAs-FUW treatment. Additionally, TNF-α/IL-4-induced inflammation in BEAS-2B cells was used as an in vitro model, whereby the effects of various doses of TAs-FUW on the TRPV1/Ca2+-dependent NFAT-induced expression of TSLP were assessed. Stimulation and inhibition of TRPV1 receptors by capsaicin (CAP) and capsazepine (CPZ), respectively, were used to validate the effect of TAs-FUW. RESULTS The UPLC-Q-TOF/MS analysis revealed that TAs-FUW mainly contain six compounds (peiminine, peimine, edpetiline, khasianine, peimisine, and sipeimine). TAs-FUW improved airway inflammation and obstruction, mucus secretion, collagen deposition, and leukocyte and macrophage infiltration, and downregulated TSLP by inhibiting the TRPV1/NFAT pathway in asthmatic mice. In vitro, the application of CPZ demonstrated that the TRPV1 channel is involved in TNF-α/IL-4-mediated regulation of TSLP. TAs-FUW suppressed TNF-α/IL-4-induced TSLP generation expression by regulating the TRPV1/Ca2+/NFAT pathway. Furthermore, TAs-FUW reduced CAP-induced TSLP release by inhibiting TRPV1 activation. Notably, sipeimine and edpetiline each were sufficient to block the TRPV1-mediated Ca2+ influx. CONCLUSION Our study is the first to demonstrate that TNF-α/IL-4 can activate the TRPV1 channel. TAs-FUW can alleviate asthmatic inflammation by suppressing the TRPV1 pathway and thereby preventing the increase in cellular Ca2+ influx and the subsequent NFAT activation. The alkaloids in FUW may be used for complementary or alternative therapies in asthma.
Collapse
Affiliation(s)
- Meihao Peng
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Jintao Li
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Jie Zhou
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Bowen Zhang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Jiaqing Liao
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Di Yang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Yu Wang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Yixi Yang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Rui Li
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Xue Tang
- Chengdu Analytical Applications Center, Shimadzu (China) Co Ltd., Chengdu 610023, China
| | - Qiuxia Lu
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Anti-infective Agent Creation Engineering Research Centre of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of pharmacy, Chengdu University, Chengdu 610106, China.
| | - Qi Zhao
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China.
| |
Collapse
|
11
|
Delbrouck JA, Desgagné M, Comeau C, Bouarab K, Malouin F, Boudreault PL. The Therapeutic Value of Solanum Steroidal (Glyco)Alkaloids: A 10-Year Comprehensive Review. Molecules 2023; 28:4957. [PMID: 37446619 DOI: 10.3390/molecules28134957] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Steroidal (glycol)alkaloids S(G)As are secondary metabolites made of a nitrogen-containing steroidal skeleton linked to a (poly)saccharide, naturally occurring in the members of the Solanaceae and Liliaceae plant families. The genus Solanum is familiar to all of us as a food source (tomato, potato, eggplant), but a few populations have also made it part of their ethnobotany for their medicinal properties. The recent development of the isolation, purification and analysis techniques have shed light on the structural diversity among the SGAs family, thus attracting scientists to investigate their various pharmacological properties. This review aims to overview the recent literature (2012-2022) on the pharmacological benefits displayed by the SGAs family. Over 17 different potential therapeutic applications (antibiotic, antiviral, anti-inflammatory, etc.) were reported over the past ten years, and this unique review analyzes each pharmacological effect independently without discrimination of either the SGA's chemical identity or their sources. A strong emphasis is placed on the discovery of their biological targets and the subsequent cellular mechanisms, discussing in vitro to in vivo biological data. The therapeutic value and the challenges of the solanum steroidal glycoalkaloid family is debated to provide new insights for future research towards clinical development.
Collapse
Affiliation(s)
- Julien A Delbrouck
- Institut de Pharmacologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Michael Desgagné
- Institut de Pharmacologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Christian Comeau
- Institut de Pharmacologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Kamal Bouarab
- Centre SEVE, Département de Biologie, Faculté des Sciences, Université de Sherbrooke, 2500 Boul de l'Université, Sherbrooke, QC J1K 2R1, Canada
| | - François Malouin
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, 2500 Boul de l'Université, Sherbrooke, QC J1K 2R1, Canada
| | - Pierre-Luc Boudreault
- Institut de Pharmacologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
12
|
Liu Y, Meng C, Li Y, Xia D, Lu C, Lai J, Zhang Y, Cao K, Gao X, Yuan Q. Peptide-Protected Gold Nanoclusters Efficiently Ameliorate Acute Contact Dermatitis and Psoriasis via Repressing the TNF-α/NF-κB/IL-17A Axis in Keratinocytes. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:662. [PMID: 36839031 PMCID: PMC9963485 DOI: 10.3390/nano13040662] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 06/18/2023]
Abstract
Immune-mediated skin diseases have a high prevalence and seriously affect patients' quality of life. Gold compounds have been considered promising therapeutic agents in dermatology, but the high incidence of adverse reactions have limited their clinical application. There is a great need to develop more effective and less toxic gold-based drugs. Gold nanoclusters fabricated by using peptides (pep-AuNCs) have appeared as potential biomedical nanomaterials because of their excellent biocompatibility, ease of fabrication and unique physicochemical properties. Glutathione (GSH) is an endogenous tripeptide and has been used for lightening the skin color. Therefore, we fabricated a well-defined gold nanocluster with GSH as an example to explore the immunomodulatory effect of AuNCs on a TNF-α-treated human keratinocyte cell line (HaCaT) in vitro, the 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced irritant contact dermatitis (ICD) model and the oxazolone (OXA)-induced psoriatic model in vivo. The results indicated that topically applied AuNCs successfully attenuated the severity of ICD and psoriasis-like lesions. In vitro and in vivo, AuNCs effectively inhibited the abnormal activation of the NF-κB pathway and the consequent overexpression of proinflammatory cytokines in keratinocytes. In particular, the transactivation of IL-17A, the most important cytokine in psoriasis pathology, was effectively inhibited by AuNCs treatment. In addition, AuNCs did not show any obvious cytotoxicity in HaCaT cells at doses even up to 100 µM and did not induce any irritation in the healthy skin and major organs, which indicated their favorable biosafety. These results indicate that biocompatible pep-AuNCs might be a promising gold-based nanomedicine for the treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- Yu Liu
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Cong Meng
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Yanggege Li
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Dongfang Xia
- College of Chemistry and Material Science, Shandong Agricultural University, Taian 271018, China
| | - Cao Lu
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Jing Lai
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Yulu Zhang
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Kai Cao
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Xueyun Gao
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Qing Yuan
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
13
|
Chen X, Zhang Y, Pei J, Zeng X, Yang Y, Zhang Y, Li F, Deng Y. Phellopterin alleviates atopic dermatitis-like inflammation and suppresses IL-4-induced STAT3 activation in keratinocytes. Int Immunopharmacol 2022; 112:109270. [PMID: 36179418 DOI: 10.1016/j.intimp.2022.109270] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022]
Abstract
Anti-inflammation medication is one of the most important treatment for people with atopic dermatitis (AD) which presents persistent type 2 inflammation in skin lesions. Interaction between activated keratinocytes and immune cells in AD skin lesions amplifies inflammatory signaling by augmenting production of cytokines, such as keratinocyte-derived thymic stromal lymphopoietin (TSLP) and interleukin-33 (IL-33). Phellopterin is a bioactive compound isolated from ethanol extract of Angelica dahurica root which has been traditionally used for AD therapy in China. In the present study, we showed that Phellopterin possessed anti-type 2 inflammation activity and alleviated AD-like phenotypes including reduction in serum immunoglobulin E (IgE) levels and infiltration of eosinophils and mast cells in the AD-like skin lesions. Further molecular analysis found that Phellopterin suppressed phosphorylation of signal transducer and activator of transcription 3 (STAT3) at Tyr705, and the expression of TSLP and IL-33 in epidermal keratinocytes of AD-like lesions. In vitro studies in cultured human keratinocytes demonstrated that STAT3 was required for interleukin-4 (IL-4)-induced overexpression of TSLP and IL-33. Phellopterin inhibited IL-4-induced activation of STAT3, which leaded to suppress the STAT3-mediated transcription of TSLP and IL-33. Our study suggested that Phellopterin is an active compound with bioactivities of anti- type 2 inflammation and STAT3 inactivation, thus allowing to be a promising candidate for AD topical therapy.
Collapse
Affiliation(s)
- Xun Chen
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Yujin Zhang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Junxia Pei
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Xin Zeng
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China; School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Yixi Yang
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China; School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - YaMei Zhang
- Key Laboratory of Clinical Genetics, Affiliated Hospital of Chengdu University, Chengdu 610106, China
| | - Fulun Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, NO. 110, Ganhe Road, Shanghai 200437, China.
| | - Yu Deng
- Engineering Research Center of Sichuan-Tibet Traditional Medicinal Plant, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China; School of Basic Medical Sciences, Chengdu University, NO. 2025, Chengluo Road, Chengdu 610106, China.
| |
Collapse
|
14
|
Li B, Liu Y, Sun S. Pump proton inhibitors display anti-tumour potential in glioma. Cell Prolif 2022:e13321. [PMID: 35961680 DOI: 10.1111/cpr.13321] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/28/2022] [Accepted: 07/14/2022] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVES Glioma is one of the most aggressive brain tumours with poor overall survival despite advanced technology in surgical resection, chemotherapy and radiation. Progression and recurrence are the hinge causes of low survival. Our aim is to explain the concrete mechanism in the proliferation and progression of tumours based on tumour microenvironment (TME). The main purpose is to illustrate the mechanism of proton pump inhibitors (PPIs) in affecting acidity, hypoxia, oxidative stress, inflammatory response and autophagy based on the TME to induce apoptosis and enhance the sensitivity of chemoradiotherapy. FINDINGS TME is the main medium for tumour growth and progression. Acidity, hypoxia, inflammatory response, autophagy, angiogenesis and so on are the main causes of tumour progress. PPIs, as a common clinical drug to inhibit gastric acid secretion, have the advantages of fast onset, long action time and small adverse reactions. Nowadays, several kinds of literature highlight the potential of PPIs in inhibiting tumour progression. However, long-term use of PPIs alone also has obvious side effects. Therefore, till now, how to apply PPIs to promote the effect of radio-chemotherapy and find the concrete dose and concentration of combined use are novel challenges. CONCLUSIONS PPIs display the potential in enhancing the sensitivity of chemoradiotherapy to defend against glioma based on TME. In the clinic, it is also necessary to explore specific concentrations and dosages in synthetic applications.
Collapse
Affiliation(s)
- Bihan Li
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Ying Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Shilong Sun
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|