1
|
Yang AY, Kim JY, Gwon MG, Kim K, Kwon HH, Leem J, Kim SW. Protective effects and mechanisms of cynaroside on renal fibrosis in mice with unilateral ureteral obstruction. Redox Rep 2025; 30:2500271. [PMID: 40322965 PMCID: PMC12054570 DOI: 10.1080/13510002.2025.2500271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
Renal fibrosis is a key factor in the progression of chronic kidney disease (CKD), and current treatments remain inadequate. In this study, we investigated the therapeutic effects of cynaroside (Cyn), a natural flavonoid, in a mouse model of renal fibrosis induced by unilateral ureteral obstruction. Cyn treatment significantly ameliorated tubular injury and interstitial fibrosis while improving renal function. Mechanistically, Cyn inhibited the expression of fibrosis-related proteins and suppressed Smad2/3 phosphorylation. Additionally, Cyn reduced myofibroblast accumulation by inhibiting epithelial-mesenchymal transition, as indicated by increased E-cadherin expression and decreased levels of mesenchymal markers. Cyn also reduced oxidative stress by downregulating the prooxidant enzyme NADPH oxidase 4 and restoring antioxidant enzymes. Furthermore, Cyn attenuated ferroptosis by regulating key proteins, including acyl-CoA synthetase long-chain family member 4, transferrin receptor 1, and glutathione peroxidase 4, while also restoring glutathione levels. Cyn alleviated endoplasmic reticulum stress, as evidenced by the downregulation of key markers such as glucose-regulated protein 78 and activating transcription factor 6, and reduced inflammation, as confirmed by decreased macrophage infiltration and lower cytokine production. Overall, Cyn demonstrated broad protective effects against renal fibrosis by modulating oxidative stress, ferroptosis, ER stress, and inflammation, positioning it as a potential therapeutic agent for CKD management.
Collapse
Affiliation(s)
- Ah Young Yang
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Jung-Yeon Kim
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Mi-Gyeong Gwon
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Kiryeong Kim
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Hyun Hee Kwon
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Sung-Woo Kim
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| |
Collapse
|
2
|
Gözcü S, Akşit Z, Aydın A, Yılmaz MA, Şimşek S. Comprehensive phenolic profiling and biological evaluation of Centaurea glastifolia L. (Asteraceae). Nat Prod Res 2025; 39:633-644. [PMID: 39267357 DOI: 10.1080/14786419.2024.2403028] [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: 03/17/2024] [Revised: 08/16/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
The present investigation focused on the comprehensive analysis of the phenolic profile of Centaurea glastifolia L. (Asteraceae) and the assessment of its diverse biological activities. Utilising LC-MS/MS, the phytochemical composition of the 70% methanol extract of Centaurea glastifolia (CG-ME) was thoroughly elucidated, revealing the presence of 30 distinct phytochemical compounds. Notably, major phenolic constituents identified in the extract included quinic acid, chlorogenic acid, luteolin-7-O-glucoside, kaempferol-3-O-glucoside, luteolin, and apigenin-7-O-glucoside. The antioxidant, antibacterial, antiproliferative, and cytotoxic activities of CG-ME were investigated. The CG-ME exhibited a moderate capacity for scavenging DPPH radicals (IC50: 50.05 ± 1.58 µg/mL) and FRAP (63.96 ± 0.39 mg TE/g extract), indicating a moderate level of antioxidant activity. Moreover, CG-ME demonstrated significant antiproliferative effects (GI50: 1.10 and 1.30 µg/mL) on cancer cells (C6 and HTC cancer cell lines, respectively) while displaying low cytotoxicity towards normal cells (LC50: >1000 µg/mL). In terms of antibacterial activity, CG-ME was found to be inactive against tested both Gram-positive and Gram-negative bacterial strains (MIC > 500 µg/mL). The extracts had a promising antiproliferative effect on C6, HeLa, and HT29 cancer cell lines with a less cytotoxic effect (10.5-14.2%) against normal cells.
Collapse
Affiliation(s)
- Sefa Gözcü
- Department of Pharmacognosy, Faculty of Pharmacy, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Zeynep Akşit
- Department of Hotel, Restaurant and Service, Tourism and Hospitality Vocational School, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Ali Aydın
- Basic Medical Science, Department Faculty of Medicine, Yozgat Bozok Universty, Yozgat, Turkey
| | - Mustafa Abdullah Yılmaz
- Department of Analytical Chemistry, Faculty of Pharmacy, Dicle University, Diyarbakır, Turkey
| | - Samed Şimşek
- Department of Medical Services and Techniques, Çayırlı Vocational School, Erzincan Binali Yıldırım University, Erzincan, Turkey
| |
Collapse
|
3
|
Ashraf M, El-Sawy HS, El Zaafarany GM, Abdel-Mottaleb MMA. Eucalyptus oil nanoemulsion for enhanced skin deposition of fluticasone propionate in psoriatic plaques: A combinatorial anti-inflammatory effect to suppress implicated cytokines. Arch Pharm (Weinheim) 2024:e2400557. [PMID: 39449230 DOI: 10.1002/ardp.202400557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
Psoriasis is a chronic inflammatory skin disease that affects patients' quality of life. This study aimed to enhance the efficacy of topical application of fluticasone propionate (FP) using a eucalyptus oil-based nanoemulsion, an oil possessing anti-inflammatory activity and extracted from the leaves, fruits, and buds of Eucalyptus globulus or Eucalyptus maidenii, to improve the skin deposition of FP and aid its anti-inflammatory effect. Box-Behnken design was employed to optimize NE formulations, which were characterized for globule size, zeta potential, polydispersity index, rheological behavior, microscopic morphology, ex vivo skin permeation/deposition, and in vivo efficacy using imiquimod-induced psoriatic lesions. The optimized formulation depicted a droplet size of 188 ± 22.4 nm, a zeta potential of -17.63 ± 1.66 mV, and a viscosity of 204.9 mPa s. In addition to the increased FP retention in different skin layers caused by the NE and the reduced PASI score compared to the marketed cream, the levels of inflammatory cytokines IL-1α, IL-6, IL17a were markedly lowered, indicating the improved anti-psoriatic curable efficacy of the optimized formulation in comparison to the FP-marketed product.
Collapse
Affiliation(s)
- Mohamed Ashraf
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Hossam S El-Sawy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
- Department of Pharmacy, Al-Kut University College, Kut, Wasit, Iraq
| | - Ghada M El Zaafarany
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mona M A Abdel-Mottaleb
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
4
|
Ebrahimi A, Mehrabi M, Miraghaee SS, Mohammadi P, Fatehi Kafash F, Delfani M, Khodarahmi R. Flavonoid compounds and their synergistic effects: Promising approaches for the prevention and treatment of psoriasis with emphasis on keratinocytes - A systematic and mechanistic review. Int Immunopharmacol 2024; 138:112561. [PMID: 38941673 DOI: 10.1016/j.intimp.2024.112561] [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: 05/09/2024] [Revised: 06/16/2024] [Accepted: 06/23/2024] [Indexed: 06/30/2024]
Abstract
Psoriasis, a chronic autoimmune skin disorder, causes rapid and excessive skin cell growth due to immune system dysfunction. Numerous studies have shown that flavonoids have anti-psoriatic effects by modulating various molecular mechanisms involved in inflammation, cytokine production, keratinocyte proliferation, and more. This study reviewed experimental data reported in scientific literature and used network analysis to identify the potential biological roles of flavonoids' targets in treating psoriasis. 947 records from Web of Sciences, ScienceDirect database, Scopus, PubMed, and Cochrane library were reviewed without limitations until June 26, 2023. 66 articles were included in the systematic review. The ten genes with the highest scores, including interleukin (IL)-10, IL-12A, IL-1β, IL-6, Tumor necrosis factor-α (TNF-α), Janus kinase 2 (JAK 2), Jun N-terminal kinase (JUN), Proto-oncogene tyrosine-protein kinase Src (SRC), Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), and Signal transducer and activator of transcription 3 (STAT3), were identified as the hub genes. KEGG pathway analysis identified connections related to inflammation and autoimmune responses, which are key characteristics of psoriasis. IL-6, STAT3, and JUN's presence in both hub and enrichment genes suggests their important role in flavonoid's effect on psoriasis. This comprehensive study highlights how flavonoids can target biological processes in psoriasis, especially when combined for enhanced effectiveness.
Collapse
Affiliation(s)
- Ali Ebrahimi
- Department of Dermatology, Hajdaie Dermatology Clinic, School of Medicine, Kermanshah University of Medical Sciences (KUMS), Kermanshah, Iran
| | - Masomeh Mehrabi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Seyyed Shahram Miraghaee
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Fatehi Kafash
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohana Delfani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
5
|
Gujarathi PP, Korat RH, Gujarathi PS. Preclinical techniques for drug discovery in psoriasis. Int Immunopharmacol 2024; 137:112378. [PMID: 38852518 DOI: 10.1016/j.intimp.2024.112378] [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: 03/02/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
Psoriasis is a chronic, inflammatory, papulosquamous, noncontagious disease characterized by scaly, demarcated erythematous plaque, affecting skin, nails, and scalp. The IL-23/Th17 axis is the main operator in the development of psoriasis. Psoriasis is affecting worldwide, and new treatment options are urgently needed. Various local and systemic treatments are available for psoriasis but they only provide symptomatic relief because of numerous unknown mechanisms. Clinical trials demand overwhelming resources; therefore, drug development predominantly depends on the in-vivo, in-vitro, and ex-vivo techniques. Immediate attention is required to develop experimental techniques that completely imitate human psoriasis to assist drug development. This review portrays the various in-vivo, in-vitro, and ex-vivo techniques used in psoriasis research. It describes these techniques' characteristics, pathological presentations, and mechanisms. The experimental techniques of psoriasis provide significant information on disease progression mechanisms and possible therapeutic targets. However, until now, it has been challenging to invent a timely, affordable model that precisely imitates a human disease. Only the xenotransplantation model is reckoned as the closer, that mimics the complete genetic, and immunopathogenic event. Imiquimod-induced psoriasis and HaCat cell lines are popular among researchers because of their convenience, ease of use, and cost-effectiveness. There need to further improve the experimental techniques to best serve the disease imitation and meet the research goal.
Collapse
Affiliation(s)
- Pranjal P Gujarathi
- Department of Pharmacology, Vidhyadeep Institute of Pharmacy, Vidhyadeep University, Anita, Surat, Gujarat, India; Bhagwan Mahavir Centre for Advance Research, Bhagwan Mahavir College of Pharmacy, Bhagwan Mahavir University, Vesu, Surat, Gujarat, India.
| | - Rashmi H Korat
- Department of Pharmacognosy, Bhagwan Mahavir College of Pharmacy, Bhagwan Mahavir Univeristy, Vesu, Surat, Gujarat, India
| | - Piyush S Gujarathi
- Department of Community Medicine, Vidhyadeep Homeopathic Medical College and Research Centre, Vidhyadeep University, Anita, Surat, Gujarat, India
| |
Collapse
|
6
|
Lee JS, Oh E, Oh H, Kim S, Ok S, Sa J, Lee JH, Shin YC, Bae YS, Choi CY, Lee S, Kwon HK, Yang S, Choi WI. Tacrolimus-loaded chitosan-based nanoparticles as an efficient topical therapeutic for the effective treatment of atopic dermatitis symptoms. Int J Biol Macromol 2024; 273:133005. [PMID: 38866268 DOI: 10.1016/j.ijbiomac.2024.133005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Atopic dermatitis (AD) is a chronic cutaneous disease with a complex underlying mechanism, and it cannot be completely cured. Thus, most treatment strategies for AD aim at relieving the symptoms. Although corticosteroids are topically applied to alleviate AD, adverse side effects frequently lead to the withdrawal of AD therapy. Tacrolimus (TAC), a calcineurin inhibitor, has been used to treat AD, but its high molecular weight and insolubility in water hinder its skin permeability. Herein, we developed and optimized TAC-loaded chitosan-based nanoparticles (TAC@CNPs) to improve the skin permeability of TAC by breaking the tight junctions in the skin. The prepared nanoparticles were highly loadable and efficient and exhibited appropriate characteristics for percutaneous drug delivery. TAC@CNP was stable for 4 weeks under physiological conditions. CNP released TAC in a controlled manner, with enhanced skin penetration observed. In vitro experiments showed that CNP was non-toxic to keratinocyte (HaCaT) cells, and TAC@CNP dispersed in an aqueous solution was as anti-proliferative as TAC solubilized in a good organic solvent. Importantly, an in vivo AD mouse model revealed that topical TAC@CNP containing ~1/10 of the dose of TAC found in commercially used Protopic® Ointment exhibited similar anti-inflammatory activity to that of the commercial product. TAC@CNP represents a potential therapeutic strategy for the management of AD.
Collapse
Affiliation(s)
- Jin Sil Lee
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea; School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Cheomdan-gwagiro, 123, Buk-gu, Gwangju 61005, Republic of Korea
| | - Eunjeong Oh
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Republic of Korea
| | - Hyeryeon Oh
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea; School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Cheomdan-gwagiro, 123, Buk-gu, Gwangju 61005, Republic of Korea
| | - Sunghyun Kim
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Subin Ok
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Junseo Sa
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | | | - Yong Chul Shin
- SKINMED Co Ltd., Daejeon 34028, Republic of Korea; Amicogen Inc, 64 Dongburo 1259, Jinsung, Jinju 52621, Republic of Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Cheol Yong Choi
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Siyoung Yang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Won Il Choi
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| |
Collapse
|
7
|
Zhao X, Dai R, Wang J, Cao L, Chen P, Yao W, Cheng F, Bao B, Zhang L. Analysis of the permeable and retainable components of Cayratia japonica ointment through intact or broken skin after topical application by UPLC-Q-TOF-MS/MS combined with in vitro transdermal assay. J Pharm Biomed Anal 2024; 238:115853. [PMID: 37976992 DOI: 10.1016/j.jpba.2023.115853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/01/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Cayratia japonica ointment has been used for many years to promote wound healing after perianal abscess surgery. This study aimed to determine the skin-permeable and skin-retainable components of Cayratia japonica ointment after topical application to intact or broken skin via UPLC-Q-TOF-MS/MS analysis and in vitro transdermal assay. Moreover, a combination of semi-quantitative and molecular docking analyses was performed to identify the main active components of the Cayratia japonica ointment and the probable phases of the wound healing process that they act on. Modified vertical Franz diffusion cells and abdominal skin of rats were selected for the in vitro transdermal study. Mass spectrometry data were collected in both positive and negative ion modes. A total of 7 flavonoids (schaftoside, luteolin-7-O-glucuronide, luteolin-7-O-glucoside, apigenin-7-O-glucuronide, luteolin, apigenin, and chrysin) and 1 coumarin (esculetin), were found to permeate and/or retained by intact or broken skin. Among them, the flavonoids were more permeable through intact/broken skin and exhibited stronger binding affinities for targets related to the inflammatory and proliferative phases of wound healing. This study suggests that the flavonoids in Cayratia japonica ointment are most likely the main active components and are crucial at the inflammatory and proliferative phases of wound healing.
Collapse
Affiliation(s)
- Xuelong Zhao
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, China
| | - Ruixue Dai
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, China
| | - Jing Wang
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Centre of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210023, Jiangsu Province, China
| | - Liangliang Cao
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, China
| | - Peidong Chen
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Centre of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210023, Jiangsu Province, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Weifeng Yao
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Centre of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210023, Jiangsu Province, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Fangfang Cheng
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Centre of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210023, Jiangsu Province, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Beihua Bao
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Centre of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210023, Jiangsu Province, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China.
| | - Li Zhang
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Centre of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210023, Jiangsu Province, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| |
Collapse
|
8
|
Myo H, Liana D, Phanumartwiwath A. Unlocking Therapeutic Potential: Comprehensive Extraction, Profiling, and Pharmacological Evaluation of Bioactive Compounds from Eclipta alba (L.) Hassk. for Dermatological Applications. PLANTS (BASEL, SWITZERLAND) 2023; 13:33. [PMID: 38202343 PMCID: PMC10781016 DOI: 10.3390/plants13010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024]
Abstract
Herbal medicine has been studied as an alternate approach to modern medicine as it is more cost-effective and accessible via natural sources. Eclipta alba (E. alba, L.) Hassk. is a weed plant abundantly distributed throughout different regions of the world and contains abundant bioactive compounds used for various skin conditions. In this review, we aimed to gather information from the literature about the extraction, separation, and identification of these bioactive compounds and their potential in skin diseases. Relevant studies published before August 2023 were identified and selected from electronic databases, including Scopus, SciFinder, ScienceDirect, Google Scholar, and Wiley Library, using the following keywords: Eclipta alba, Eclipta prostrata, phytochemicals, extraction, separation, isolation, identification, characterization, pharmacological activity, and skin conditions. Up-to-date extraction, separation, and identification methods of bioactive compounds from E. alba and their skin-related pharmacological activities are discussed in this review. As there are limitations regarding extraction, separation, and identification methods, and in-depth mechanistic and human studies of the skin-related pharmacological activities of bioactive compounds, these gaps are areas for future research to expand our understanding and broaden the potential applications of this medicinal weed plant, including the development of cosmeceutical and skincare products, anti-inflammatory agents, and formulations for dermatological treatments.
Collapse
Affiliation(s)
| | | | - Anuchit Phanumartwiwath
- College of Public Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (H.M.); (D.L.)
| |
Collapse
|
9
|
Gyuzeleva D, Benina M, Ivanova V, Vatov E, Alseekh S, Mladenova T, Mladenov R, Todorov K, Bivolarska A, Stoyanov P. Metabolome Profiling of Marrubium peregrinum L. and Marrubium friwaldskyanum Boiss Reveals Their Potential as Sources of Plant-Based Pharmaceuticals. Int J Mol Sci 2023; 24:17035. [PMID: 38069358 PMCID: PMC10707198 DOI: 10.3390/ijms242317035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Marrubium species have been used since ancient times as food additives and curative treatments. Their phytochemical composition and various pharmacological activities were the focus of a number of scientific investigations but no comprehensive metabolome profiling to identify the numerous primary and secondary metabolites has been performed so far. This study aimed to generate a comprehensive picture of the total metabolite content of two Marrubium species-M. peregrinum and M. friwaldskyanum-to provide detailed information about the main primary and secondary metabolites. In addition, the elemental composition was also evaluated. For this purpose, non-targeted metabolomic analyses were conducted using GC-MS, UPLC-MS/MS and ICP-MS approaches. Nearly 500 compounds and 12 elements were detected and described. The results showed a strong presence of phenolic acids, flavonoids and their glucosides, which are generally of great interest due to their various pharmacological activities. Furthermore, tissue-specific analyses for M. friwaldskyanum stem, leaves and flowers were carried out in order to outline the sources of potentially important bioactive molecules. The results generated from this study depict the Marrubium metabolome and reveal its dual scientific importance-from one side, providing information about the metabolites that is fundamental and vital for the survival of these species, and from the other side, defining the large diversity of secondary substances that are a potential source of phytotherapeutic agents.
Collapse
Affiliation(s)
- Donika Gyuzeleva
- Department of Botany and Biological Education, Faculty of Biology, University of Plovdiv “Paisii Hilendarski”, 24 Tsar Assen Str., 4000 Plovdiv, Bulgaria; (D.G.); (T.M.)
| | - Maria Benina
- Center of Plant Systems Biology and Biotechnology, 14 Sveti Kniaz Boris I Pokrastitel Str., 4023 Plovdiv, Bulgaria
| | - Valentina Ivanova
- Center of Plant Systems Biology and Biotechnology, 14 Sveti Kniaz Boris I Pokrastitel Str., 4023 Plovdiv, Bulgaria
| | - Emil Vatov
- Center of Plant Systems Biology and Biotechnology, 14 Sveti Kniaz Boris I Pokrastitel Str., 4023 Plovdiv, Bulgaria
| | - Saleh Alseekh
- Center of Plant Systems Biology and Biotechnology, 14 Sveti Kniaz Boris I Pokrastitel Str., 4023 Plovdiv, Bulgaria
- Max Planck Institute for Molecular Plant Physiology, 1, Am Mühlenberg, 14476 Potsdam, Germany
| | - Tsvetelina Mladenova
- Department of Botany and Biological Education, Faculty of Biology, University of Plovdiv “Paisii Hilendarski”, 24 Tsar Assen Str., 4000 Plovdiv, Bulgaria; (D.G.); (T.M.)
| | - Rumen Mladenov
- Department of Botany and Biological Education, Faculty of Biology, University of Plovdiv “Paisii Hilendarski”, 24 Tsar Assen Str., 4000 Plovdiv, Bulgaria; (D.G.); (T.M.)
- Department of Bioorganic Chemistry, Faculty of Pharmacy, Medical University of Plovdiv, 15A Vasil Aprilov Blvd., 4002 Plovdiv, Bulgaria
| | - Krasimir Todorov
- Department of Botany and Biological Education, Faculty of Biology, University of Plovdiv “Paisii Hilendarski”, 24 Tsar Assen Str., 4000 Plovdiv, Bulgaria; (D.G.); (T.M.)
| | - Anelia Bivolarska
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, 15A Vasil Aprilov Blvd., 4002 Plovdiv, Bulgaria
| | - Plamen Stoyanov
- Department of Botany and Biological Education, Faculty of Biology, University of Plovdiv “Paisii Hilendarski”, 24 Tsar Assen Str., 4000 Plovdiv, Bulgaria; (D.G.); (T.M.)
- Department of Bioorganic Chemistry, Faculty of Pharmacy, Medical University of Plovdiv, 15A Vasil Aprilov Blvd., 4002 Plovdiv, Bulgaria
| |
Collapse
|
10
|
Rakoczy K, Kaczor J, Sołtyk A, Szymańska N, Stecko J, Sleziak J, Kulbacka J, Baczyńska D. Application of Luteolin in Neoplasms and Nonneoplastic Diseases. Int J Mol Sci 2023; 24:15995. [PMID: 37958980 PMCID: PMC10650338 DOI: 10.3390/ijms242115995] [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: 09/24/2023] [Revised: 10/31/2023] [Accepted: 11/04/2023] [Indexed: 11/15/2023] Open
Abstract
Researchers are amazed at the multitude of biological effects of 3',4',5,7-tetrahydroxyflavone, more commonly known as luteolin, as it simultaneously has antioxidant and pro-oxidant, as well as antimicrobial, anti-inflammatory, and cancer-preventive, properties. The anticancer properties of luteolin constitute a mosaic of pathways due to which this flavonoid influences cancer cells. Not only is it able to induce apoptosis and inhibit cancer cell proliferation, but it also suppresses angiogenesis and metastasis. Moreover, luteolin succeeds in cancer cell sensitization to therapeutically induced cytotoxicity. Nevertheless, apart from its promising role in chemoprevention, luteolin exhibits numerous potential utilizations in patients with conditions other than neoplasms, which include inflammatory skin diseases, diabetes mellitus, and COVID-19. This review aims to present the multidimensionality of the luteolin's impact on both neoplastic and nonneoplastic diseases. When it comes to neoplasms, we intend to describe the complexity of the molecular mechanisms that underlay luteolin's anticancer effectiveness, as well as to prove the usefulness of integrating this flavonoid in cancer therapy via the analysis of recent research on breast, colon, and lung cancer. Regarding nonneoplastic diseases, this review aims to emphasize the importance of researching the potential of luteolin in areas such as diabetology, virology, and dermatology as it summarizes the most important discoveries in those fields regarding its application.
Collapse
Affiliation(s)
- Katarzyna Rakoczy
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.); (J.S.)
| | - Justyna Kaczor
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.); (J.S.)
| | - Adam Sołtyk
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.); (J.S.)
| | - Natalia Szymańska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.); (J.S.)
| | - Jakub Stecko
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.); (J.S.)
| | - Jakub Sleziak
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.); (J.S.)
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, 08410 Vilnius, Lithuania
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
11
|
Dah-Nouvlessounon D, Chokki M, Agossou EA, Houédanou JB, Nounagnon M, Sina H, Vulturar R, Heghes SC, Cozma A, Mavoungou JF, Fodor A, Baba-Moussa F, Suharoschi R, Baba-Moussa L. Polyphenol Analysis via LC-MS-ESI and Potent Antioxidant, Anti-Inflammatory, and Antimicrobial Activities of Jatropha multifida L. Extracts Used in Benin Pharmacopoeia. Life (Basel) 2023; 13:1898. [PMID: 37763302 PMCID: PMC10532662 DOI: 10.3390/life13091898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/08/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Jatropha multifida L., a plant from the Euphorbiaceae family, is commonly used in Benin's traditional medicine due to its therapeutic benefits. This study aims to explore the medicinal efficacy of Jatropha multifida L. by evaluating its various biological activities. An initial phytochemical analysis was conducted, following which the polyphenols and flavonoids were quantified and identified using LC-MS-ESI. The antimicrobial efficacy of the extracts was tested using agar diffusion. Their antioxidant capacity was assessed using several methods: DPPH radical reduction, ABTS radical cation reduction, ferric ion (FRAP) reduction, and lipid peroxidation (LPO). Anti-inflammatory activity was determined based on the inhibition of protein (specifically albumin) denaturation. The study identified several phenolic and flavonoid compounds, including 2-Hydroxybenzoic acid, o-Coumaroylquinic acid, Apigenin-apiosyl-glucoside, and luteolin-galactoside. Notably, the extracts of J. multifida demonstrated bactericidal effects against a range of pathogens, with Concentration Minimally Bactericidal (CMB) values ranging from 22.67 mg/mL (for organisms such as S. aureus and C. albicans) to 47.61 mg/mL (for E. coli). Among the extracts, the ethanolic variant displayed the most potent DPPH radical scavenging activity, with an IC50 value of 0.72 ± 0.03 mg/mL. In contrast, the methanolic extract was superior in ferric ion reduction, registering 46.23 ± 1.10 µgEAA/g. Interestingly, the water-ethanolic extract surpassed others in the ABTS reduction method with a score of 0.49 ± 0.11 mol ET/g and also showcased the highest albumin denaturation inhibition rate of 97.31 ± 0.35% at a concentration of 1000 µg/mL. In conclusion, the extracts of Jatropha multifida L. are enriched with bioactive compounds that exhibit significant biological activities, underscoring their therapeutic potential.
Collapse
Affiliation(s)
- Durand Dah-Nouvlessounon
- Laboratory of Biology and Molecular Typing in Microbiology, Department of Biochemistry and Cell Biology, Faculty of Sciences and Techniques, University of Abomey-Calavi, Cotonou 05BP1604, Benin; (D.D.-N.); (J.-B.H.); (M.N.); (H.S.)
| | - Michaelle Chokki
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 3-5 Calea Manastur Street, 400372 Cluj-Napoca, Romania;
- Laboratoire de Microbiologie et de Technologie Alimentaire, FAST, Université d’Abomey-Calavi, 01BP: 526 ISBA-Champ de Foire, Cotonou 01BP188, Benin;
| | - Essé A. Agossou
- Laboratory of Pharmacology and Improved Traditional Medicines, FAST, Department of Animal Physiology, University of Abomey-Calavi, Cotonou 01BP526, Benin;
| | - Jean-Baptiste Houédanou
- Laboratory of Biology and Molecular Typing in Microbiology, Department of Biochemistry and Cell Biology, Faculty of Sciences and Techniques, University of Abomey-Calavi, Cotonou 05BP1604, Benin; (D.D.-N.); (J.-B.H.); (M.N.); (H.S.)
| | - Martial Nounagnon
- Laboratory of Biology and Molecular Typing in Microbiology, Department of Biochemistry and Cell Biology, Faculty of Sciences and Techniques, University of Abomey-Calavi, Cotonou 05BP1604, Benin; (D.D.-N.); (J.-B.H.); (M.N.); (H.S.)
| | - Haziz Sina
- Laboratory of Biology and Molecular Typing in Microbiology, Department of Biochemistry and Cell Biology, Faculty of Sciences and Techniques, University of Abomey-Calavi, Cotonou 05BP1604, Benin; (D.D.-N.); (J.-B.H.); (M.N.); (H.S.)
| | - Romana Vulturar
- Department of Molecular Sciences, “luliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes, 400012 Cluj-Napoca, Romania;
| | - Simona Codruta Heghes
- Department of Drug Analysis, “luliu Hatieganu” University of Medicine and Pharmacy, 6 Louis Pasteur Street, 400012 Cluj-Napoca, Romania;
| | - Angela Cozma
- Internal Medicine Department, 4th Medical Clinic “luliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Jacques François Mavoungou
- Department of Microbiology, International University of Libreville, ESSASSA-Libreville Campus, Essassa BP 20411, Gabon;
| | - Adriana Fodor
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “luliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Farid Baba-Moussa
- Laboratoire de Microbiologie et de Technologie Alimentaire, FAST, Université d’Abomey-Calavi, 01BP: 526 ISBA-Champ de Foire, Cotonou 01BP188, Benin;
| | - Ramona Suharoschi
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 3-5 Calea Manastur Street, 400372 Cluj-Napoca, Romania;
| | - Lamine Baba-Moussa
- Laboratory of Biology and Molecular Typing in Microbiology, Department of Biochemistry and Cell Biology, Faculty of Sciences and Techniques, University of Abomey-Calavi, Cotonou 05BP1604, Benin; (D.D.-N.); (J.-B.H.); (M.N.); (H.S.)
| |
Collapse
|
12
|
Fu D, Zheng S, Li J, Hu H, Wang Q, Fu X, Li M, Yan D, Yang Z, Tian Z, Song X. Anti-interleukin 33 treatment alleviates psoriatic dermatitis in mice induced imiquimod. Int Immunopharmacol 2023; 121:110480. [PMID: 37343370 DOI: 10.1016/j.intimp.2023.110480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/23/2023]
Abstract
Interleukin-33(IL-33), is constitutively expressed in the epithelial cells of the skin. It has been reported that IL-33 contributed to the severity of the disease in psoriasis-like mouse models. In the current study, we evaluated the effect of anti-IL-33 antibody (Ab) in imiquimod-induced psoriatic dermatitis in mice. Our observations showed that anti-IL-33 Ab ameliorated the erythema, scaling, epidermal thickness and spleen index. Additionally, we found anti-IL-33 Ab significantly decreased the expression of psoriasis-related cytokines. Moreover, anti-IL-33 Ab significantly reduced Ki-67 positive cells, CD3+CD4+T cells, and CD3+CD8+T cells in the skin lesions. Furthermore, anti-IL-33 Ab treatment down-regulated the expression of phosphorylation of STAT3 and IL-33 in model mouse. These results indicated that the anti-IL-33 Ab alleviated the seriousness of skin lesions, inhibited the activation of the STAT3, lymphocyte infiltration and the secretion of pro-inflammatory cytokines in imiquimod-induced psoriatic dermatitis in mice, suggesting IL-33 may be a therapeutic target for the treatment of psoriasis.
Collapse
Affiliation(s)
- Dandan Fu
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Shuting Zheng
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Jialin Li
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Hua Hu
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Qingqing Wang
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Xiuyu Fu
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Min Li
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Dong Yan
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Zishan Yang
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453000, Henan, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, China
| | - Zhongwei Tian
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China.
| | - Xiangfeng Song
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453000, Henan, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, China.
| |
Collapse
|
13
|
Marques MP, Varela C, Mendonça L, Cabral C. Nanotechnology-Based Topical Delivery of Natural Products for the Management of Atopic Dermatitis. Pharmaceutics 2023; 15:1724. [PMID: 37376172 DOI: 10.3390/pharmaceutics15061724] [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: 05/05/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic eczematous inflammatory disease that may arise from environmental, genetic, and immunological factors. Despite the efficacy of current treatment options such as corticosteroids, such approaches are mainly focused on symptom relief and may present certain undesirable side effects. In recent years, isolated natural compounds, oils, mixtures, and/or extracts have gained scientific attention because of their high efficiency and moderate to low toxicity. Despite their promising therapeutic effects, the applicability of such natural healthcare solutions is somewhat limited by their instability, poor solubility, and low bioavailability. Therefore, novel nanoformulation-based systems have been designed to overcome these limitations, thus enhancing the therapeutic potential, by promoting the capacity of these natural drugs to properly exert their action in AD-like skin lesions. To the best of our knowledge, this is the first literature review that has focused on summarizing recent nanoformulation-based solutions loaded with natural ingredients, specifically for the management of AD. We suggest that future studies should focus on robust clinical trials that may confirm the safety and effectiveness of such natural-based nanosystems, thus paving the way for more reliable AD treatments.
Collapse
Affiliation(s)
- Mário Pedro Marques
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Carla Varela
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Chemical Process Engineering and Forest Products (CIEPQPF), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Laura Mendonça
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Centre for Functional Ecology, Department of Life Sciences, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Célia Cabral
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Centre for Functional Ecology, Department of Life Sciences, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
14
|
Sawa T, Endo K, Nakashima A, Tanaka Y, Makabe H, Tanaka S. Procyanidin B2 3,3''-di-O-gallate ameliorates imiquimod-induced skin inflammation by suppressing TLR7 signaling through the inhibition of endosomal acidification in dendritic cells. Int Immunopharmacol 2023; 121:110444. [PMID: 37295030 DOI: 10.1016/j.intimp.2023.110444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/11/2023]
Abstract
The excessive activation of abnormal T helper 17 (Th17) cells and dendritic cells (DCs) in the dermis and epidermis causes severe inflammation of the skin. Toll-like receptor 7 (TLR7)-located in the endosomes of DCs-recognizes nucleic acids from pathogens as well as imiquimod (IMQ), which plays a crucial role in the pathogenesis of skin inflammation. Procyanidin B2 3,3''-di-O-gallate (PCB2DG), a polyphenol, has been reported to suppress the excessive production of proinflammatory cytokines from T cells. The aim of this study was to demonstrate the inhibitory effect of PCB2DG on skin inflammation and TLR7 signaling in DCs. In vivo studies showed that the clinical symptoms of dermatitis were markedly improved by the oral administration of PCB2DG in mouse dermatitis model caused by IMQ application, accompanied by the suppression of excessive cytokine secretion in the inflamed skin and spleen. In vitro, PCB2DG significantly decreased cytokine production in TLR7- or TLR9 ligand-stimulated bone marrow-derived dendritic cells (BMDCs), suggesting that PCB2DG suppresses endosomal toll-like receptors (TLR) signaling in DCs. The activity of endosomal TLRs depends on endosomal acidification, which was significantly inhibited by PCB2DG in BMDCs. The addition of cAMP, an accelerator of endosomal acidification, abrogated the inhibitory effect of cytokine production by PCB2DG. These results provide a new insight into developing functional foods, including PCB2DG, to improve the symptoms of skin inflammation through the suppression of TLR7 signaling in DCs.
Collapse
Affiliation(s)
- Toko Sawa
- Division of Food Science and Biotechnology, Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Katsunori Endo
- Division of Food Science and Biotechnology, Department of Science and Technology Agriculture, Graduate School of Medicine, Science and Technology, Shinshu University, Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Akane Nakashima
- Department of Agricultural and Life Science, Faculty of Agriculture, Shinshu University, Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Yuna Tanaka
- Division of Food Science and Biotechnology, Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Hidefumi Makabe
- Division of Food Science and Biotechnology, Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Minami-minowa, Kami-ina, Nagano 399-4598, Japan; Division of Food Science and Biotechnology, Department of Science and Technology Agriculture, Graduate School of Medicine, Science and Technology, Shinshu University, Minami-minowa, Kami-ina, Nagano 399-4598, Japan; Department of Agricultural and Life Science, Faculty of Agriculture, Shinshu University, Minami-minowa, Kami-ina, Nagano 399-4598, Japan; Division of Innovative Biomolecular Science, Interdisciplinary Cluster for Cutting Edge, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa Kami-ina, Nagano 399-4598, Japan
| | - Sachi Tanaka
- Division of Food Science and Biotechnology, Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Minami-minowa, Kami-ina, Nagano 399-4598, Japan; Division of Food Science and Biotechnology, Department of Science and Technology Agriculture, Graduate School of Medicine, Science and Technology, Shinshu University, Minami-minowa, Kami-ina, Nagano 399-4598, Japan; Department of Agricultural and Life Science, Faculty of Agriculture, Shinshu University, Minami-minowa, Kami-ina, Nagano 399-4598, Japan.
| |
Collapse
|
15
|
Bernal-Millán MDJ, Carrasco-Portugal MDC, Heredia JB, Bastidas-Bastidas PDJ, Gutiérrez-Grijalva EP, León-Félix J, Angulo-Escalante MÁ. Green Extracts and UPLC-TQS-MS/MS Profiling of Flavonoids from Mexican Oregano ( Lippia graveolens) Using Natural Deep Eutectic Solvents/Ultrasound-Assisted and Supercritical Fluids. PLANTS (BASEL, SWITZERLAND) 2023; 12:1692. [PMID: 37111915 PMCID: PMC10145289 DOI: 10.3390/plants12081692] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/14/2023] [Accepted: 04/16/2023] [Indexed: 06/19/2023]
Abstract
Mexican oregano (Lippia graveolens) is an important source of bioactive compounds, such as flavonoids. These have presented different therapeutic properties, including antioxidant and anti-inflammatory; however, their functionality is related to the quantity and type of compounds, and these characteristics depend on the extraction method used. This study aimed to compare different extraction procedures to identify and quantify flavonoids from oregano (Lippia graveolens). Emerging and conventional technologies include maceration with methanol and water, and ultrasound-assisted extraction (UAE) using deep eutectic solvents (DES) such as choline chloride-ethylene glycol, choline chloride-glycerol, and choline chloride-lactic acid. Supercritical fluid extraction using CO2 as a solvent was also studied. Six different extracts were obtained and the total reducing capacity, total flavonoid content, and antioxidant capacity by ABTS•+, DPPH•, FRAP, and ORAC were evaluated. In addition, flavonoids were identified and quantified by UPLC-TQS-MS/MS. Results showed that UAE-DES had the best extraction effect and antioxidant capacity using colorimetric methods. However, maceration-methanol was superior in compound content, and highlighting naringenin and phloridzin were the major compounds. In addition, this extract was microencapsulated by spray drying, which provided a protection feature of their antioxidant potential. Oregano extracts are rich in flavonoids and the microcapsules present promising results for future research.
Collapse
Affiliation(s)
| | - Miriam del Carmen Carrasco-Portugal
- Unidad de Investigación en Farmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México 14080, Mexico
| | - J. Basilio Heredia
- Centro de Investigación en Alimentación y Desarrollo A.C., Culiacán 80110, Mexico; (M.d.J.B.-M.)
| | | | | | - Josefina León-Félix
- Centro de Investigación en Alimentación y Desarrollo A.C., Culiacán 80110, Mexico; (M.d.J.B.-M.)
| | | |
Collapse
|
16
|
Can Essential Oils/Botanical Agents Smart-Nanoformulations Be the Winning Cards against Psoriasis? Pharmaceutics 2023; 15:pharmaceutics15030750. [PMID: 36986611 PMCID: PMC10056241 DOI: 10.3390/pharmaceutics15030750] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 02/26/2023] Open
Abstract
Although psoriasis remains one of the most devastating inflammatory disorders due to its huge negative impact on patients’ quality of life, new “green” treatment approaches still need to be fully explored. The purpose of this review article is to focus on the utilization of different essential oils and active constituents of herbal botanical origin for the treatment of psoriasis that proved efficacious via both in vitro and in vivo models. The applications of nanotechnology-based formulations which displayed great potential in augmenting the permeation and delivery of these agents is also addressed. Numerous studies have been found which assessed the potential activity of natural botanical agents to overcome psoriasis. Nano-architecture delivery is applied in order to maximize the benefits of their activity, improve properties, and increase patient compliance. This field of natural innovative formulations can be a promising tool to optimize remediation of psoriasis while minimizing adverse effects.
Collapse
|
17
|
Kumar A, Kaur S, Sangwan PL, Tasduq SA. Therapeutic and cosmeceutical role of glycosylated natural products in dermatology. Phytother Res 2023; 37:1574-1589. [PMID: 36809543 DOI: 10.1002/ptr.7752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 09/03/2022] [Accepted: 10/09/2022] [Indexed: 02/23/2023]
Abstract
Natural products (NPs) remain the primary source of pharmacologically active candidates for drug discovery. Since time immemorial, NPs have attracted considerable attention because of their beneficial skin effects. Moreover, there has been a great interest in using such products for the cosmetics industry in the past few decades, bridging the gap between modern and traditional medicine. Terpenoids, Steroids, and Flavonoids having glycosidic attachment have proven biological effects with a positive impact on human health. NPs derived glycosides are mainly found in fruits, vegetables, and plants, and most of them have a special reverence in traditional and modern medicine for disease prevention and treatment. A literature review was performed using scientific journals, Google scholar, Scifinder, PubMED, and Google patents. These scientific articles, documents, and patents establish the significance of glycosidic NPs in the areas of dermatology. Considering the human inclination to the usage of NPs rather than synthetic or inorganic drugs (especially in the area of skin care), in the present review we have discussed the worth of NP glycosides in beauty care and skin-related therapeutics and the mechanistic pathways involved.
Collapse
Affiliation(s)
- Amit Kumar
- Natural Product and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India.,PK/PD divisions, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Sarabjit Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Pyare L Sangwan
- Natural Product and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Sheikh A Tasduq
- PK/PD divisions, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,PK-PD and Toxicology Divisions, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
18
|
Aruncus dioicus var. kamtschaticus Extract Ameliorates Psoriasis-like Skin Inflammation via Akt/mTOR and JAK2/STAT3 Signaling Pathways in a Murine Model. Nutrients 2022; 14:nu14235094. [PMID: 36501124 PMCID: PMC9736163 DOI: 10.3390/nu14235094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Goat's beard (Aruncus dioicus var. kamtschaticus) is a traditional medicinal plant, widely used in Chinese and Korean traditional medicine because of its anti-inflammatory, anti-oxidant, antimicrobial, and anti-cancer activity. However, its effect on skin inflammatory diseases like psoriasis is unknown. The aim of this study was to investigate the therapeutic potency of A. dioicus extract (ADE) in in vitro and in vivo psoriasis models. ADE treatment significantly attenuated skin inflammation and improved skin integrity in imiquimod-treated mice by suppressing keratinocyte hyperproliferation, inhibiting the infiltration of immune cells, and downregulating the expression of psoriatic markers. Further, ADE treatment suppressed protein kinase B/mammalian target of rapamycin (Akt/mTOR) and Janus kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT3) signaling in HaCaT cells. Overall, the application of ADE relieves psoriasis-like skin inflammation possibly by regulating the Akt/mTOR and JAK2/STAT3 signaling pathways, making it an effective alternative for psoriasis therapy.
Collapse
|
19
|
Xi L, Han Y, Liu C, Liu Y, Wang Z, Wang R, Zheng Y. Sonodynamic therapy by phase-transition nanodroplets for reducing epidermal hyperplasia in psoriasis. J Control Release 2022; 350:435-447. [PMID: 36030991 DOI: 10.1016/j.jconrel.2022.08.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/31/2022] [Accepted: 08/21/2022] [Indexed: 11/29/2022]
Abstract
The cross-talk between hyperproliferative keratinocytes and activated immune cells is responsible for the progression of psoriasis. The strategy to alleviate psoriasis through inhibiting the abnormal proliferation of keratinocytes remains challenging due to limited therapeutic effects and low skin penetration of drugs. Herein we designed an ultrasound-triggered phase-transition nanodroplet that could produce cavitation to enhance skin penetration and effectively generate reactive oxygen species (ROS) to induce keratinocyte apoptosis for psoriasis treatment. After ultrasound stimulation, the perfluoro-n-pentane (PFP) liquid core of the nanodroplets vaporized, and the Haematoporphyrin monomethyl ether (HMME) encapsulated in the nanodroplets generated plenty of intracellular ROS which caused the apoptosis of HaCat cells through inducing mitochondrial dysfunction. In addition, the blank nanodroplets successfully inhibited the secretion of IL-6 and TNF-α from macrophages and dendritic cells in vitro due to the anti-inflammatory effect of POPG. For the skin penetration test, the phase-transition nanodroplets could effectively accumulate in the epidermis of the skin and generate intracellular ROS. The in-vivo anti-psoriasis experiment demonstrated that the phase-transition nanodroplets relieved the symptoms of psoriasis lesion and inhibited epidermal hyperplasia through induction of cell apoptosis under ultrasound irritation. Meanwhile, the inflammatory cytokines in the skin lesion almost decreased to the normal baseline level after SDT. Collectively, this study demonstrated a new strategy to inhibit keratinocyte hyperproliferation for psoriasis management based on sonodynamic responded nanodroplets.
Collapse
Affiliation(s)
- Long Xi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Yunfeng Han
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Chang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Yihan Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, CA 92093, USA
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China.
| |
Collapse
|
20
|
Li T, Gao S, Han W, Gao Z, Wei Y, Wu G, Qiqiu W, Chen L, Feng Y, Yue S, Kuang H, Jiang X. Potential effects and mechanisms of Chinese herbal medicine in the treatment of psoriasis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 294:115275. [PMID: 35487447 DOI: 10.1016/j.jep.2022.115275] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/17/2022] [Accepted: 04/05/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Psoriasis is a chronic inflammatory dermatosis related to high morbidity and mortality. The incidence of psoriasis is increasing in recent decades. Some patients with psoriasis are anxious about the underlying side effects of synthetic drugs they are on. Therefore, they are eager to seek alternative and efficient therapy, such as Chinese herbal medicine (CHM). Researchers have found some CHM provides best source for the development of anti-psoriatic drugs because of their structural diversity and fewer adverse reactions. Some of CHM formulas or active constituents extracted from CHM have been rapidly developed into clinical drugs with good efficacy. At present, along with the CHM formulas, single CHM and its active components have been extensively accepted and utilized in the treatment of psoriasis, whose therapeutic mechanisms hitherto have not been thoroughly illustrated. AIM OF THE STUDY This review aimed to comprehensively summarize about the existing therapeutic mechanisms of CHM in the treatment of psoriasis and to provide a reference to develop future related studies in this field. MATERIALS AND METHODS Relevant literatures about how CHM treated psoriasis were acquired from published scientific studies (including PubMed, CNKI, Web of Science, Baidu Scholar, The Plant List, Elsevier and SciFinder). All plants appearing in the review have been included in The Plant List or Medicinal Plant Names Services (MPNS). RESULTS In this review, we collect numerous literatures about how CHM treats psoriasis via immune cells, signaling pathways and disease-related mediators and systematically elucidates potential mechanisms from the point of the suppression of oxidative stress, the inhibition of abnormal abnormal proliferation and differentiation, the inhibition of immune responses, and the suppression of angiogenesis. CONCLUSIONS Psoriasis is considered as a complicated disease caused by interaction among various mechanisms. The CHM formulas, single CHM and its active components have considerable positive reports about the treatment of psoriasis, which brings hope for a promising future of CHM in the clinical therapy of psoriasis. In the paper, we have concluded that the existing therapeutic mechanisms of CHM in the treatment of psoriasis.
Collapse
Affiliation(s)
- Tingting Li
- School of Medicine, Guangxi University of Science and Technology, No.257 Liu-shi Road, Yufeng District, Liuzhou, 545005, China
| | - Si Gao
- School of Medicine, Guangxi University of Science and Technology, No.257 Liu-shi Road, Yufeng District, Liuzhou, 545005, China
| | - Wei Han
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, No.4 Dong-qing Road, Huaxi District, Guiyang, 550025, China
| | - Zhenqiu Gao
- School of Pharmacy, Yancheng Teachers University, Xiwang Road, Tinghu District, Yancheng, 224007, China
| | - Yundong Wei
- School of Medicine, Guangxi University of Science and Technology, No.257 Liu-shi Road, Yufeng District, Liuzhou, 545005, China
| | - Gang Wu
- School of Medicine, Guangxi University of Science and Technology, No.257 Liu-shi Road, Yufeng District, Liuzhou, 545005, China
| | - Wei Qiqiu
- School of Medicine, Guangxi University of Science and Technology, No.257 Liu-shi Road, Yufeng District, Liuzhou, 545005, China
| | - Li Chen
- School of Medicine, Guangxi University of Science and Technology, No.257 Liu-shi Road, Yufeng District, Liuzhou, 545005, China
| | - Yiping Feng
- School of Medicine, Guangxi University of Science and Technology, No.257 Liu-shi Road, Yufeng District, Liuzhou, 545005, China
| | - Shijiao Yue
- Gangnan Castle Peak Psychiatric Hospital, Jiangnan Industrial Park District, Guigang, 537100, China
| | - Haixue Kuang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Traditional Chinese Medicine, No.24 Heping Road, Xiangfang District, Harbin, 150040, China.
| | - Xudong Jiang
- School of Medicine, Guangxi University of Science and Technology, No.257 Liu-shi Road, Yufeng District, Liuzhou, 545005, China.
| |
Collapse
|
21
|
Characterization of Constituents with Potential Anti-Inflammatory Activity in Chinese Lonicera Species by UHPLC-HRMS Based Metabolite Profiling. Metabolites 2022; 12:metabo12040288. [PMID: 35448474 PMCID: PMC9027581 DOI: 10.3390/metabo12040288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/16/2022] [Accepted: 03/19/2022] [Indexed: 01/02/2023] Open
Abstract
This study centered on detecting potentially anti-inflammatory active constituents in ethanolic extracts of Chinese Lonicera species by taking an UHPLC-HRMS-based metabolite profiling approach. Extracts from eight different Lonicera species were subjected to both UHPLC-HRMS analysis and to pharmacological testing in three different cellular inflammation-related assays. Compounds exhibiting high correlations in orthogonal projections to latent structures discriminant analysis (OPLS-DA) of pharmacological and MS data served as potentially activity-related candidates. Of these candidates, 65 were tentatively or unambiguously annotated. 7-Hydroxy-5,3′,4′,5′-tetramethoxyflavone and three bioflavonoids, as well as three C32- and one C34-acetylated polyhydroxy fatty acid, were isolated from Lonicera hypoglauca leaves for the first time, and their structures were fully or partially elucidated. Of the potentially active candidate compounds, 15 were subsequently subjected to pharmacological testing. Their activities could be experimentally verified in part, emphasizing the relevance of Lonicera species as a source of anti-inflammatory active constituents. However, some compounds also impaired the cell viability. Overall, the approach was found useful to narrow down the number of potentially bioactive constituents in the complex extracts investigated. In the future, the application of more refined concepts, such as extract prefractionation combined with bio-chemometrics, may help to further enhance the reliability of candidate selection.
Collapse
|
22
|
Anti-Inflammatory and Active Biological Properties of the Plant-Derived Bioactive Compounds Luteolin and Luteolin 7-Glucoside. Nutrients 2022; 14:nu14061155. [PMID: 35334812 PMCID: PMC8949538 DOI: 10.3390/nu14061155] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/02/2022] [Accepted: 03/05/2022] [Indexed: 12/14/2022] Open
Abstract
Flavonoids are interesting molecules synthetized by plants. They can be found abundantly in seeds and fruits, determining the color, flavor, and other organoleptic characteristics, as well as contributing to important nutritional aspects. Beyond these characteristics, due to their biochemical properties and characteristics, they can be considered bioactive compounds. Several interesting studies have demonstrated their biological activity in different cellular and physiological processes in high-order organisms including humans. The flavonoid molecular structure confers the capability of reacting with and neutralizing reactive oxygen species (ROS), behaving as scavengers in all processes generating this class of molecules, such as UV irradiation, a process widely present in plant physiology. Importantly, the recent scientific literature has demonstrated that flavonoids, in human physiology, are active compounds acting not only as scavengers but also with the important role of counteracting the inflammation process. Among the wide variety of flavonoid molecules, significant results have been shown by investigating the role of the flavones luteolin and luteolin-7-O-glucoside (LUT-7G). For these compounds, experimental results demonstrated an interesting anti-inflammatory action, both in vitro and in vivo, in the interaction with JAK/STAT3, NF-κB, and other pathways described in this review. We also describe the effects in metabolic pathways connected with inflammation, such as cellular glycolysis, diabetes, lipid peroxidation, and effects in cancer cells. Moreover, the inhibition of inflammatory pathway in endothelial tissue, as well as the NLRP3 inflammasome assembly, demonstrates a key role in the progression of such phenomena. Since these micronutrient molecules can be obtained from food, their biochemical properties open new perspectives with respect to the long-term health status of healthy individuals, as well as their use as a coadjutant treatment in specific diseases.
Collapse
|
23
|
Neuroprotective Effect of Luteolin-7-O-Glucoside against 6-OHDA-Induced Damage in Undifferentiated and RA-Differentiated SH-SY5Y Cells. Int J Mol Sci 2022; 23:ijms23062914. [PMID: 35328335 PMCID: PMC8949357 DOI: 10.3390/ijms23062914] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
Luteolin is one of the most common flavonoids present in edible plants and its potential benefits to the central nervous system include decrease of microglia activation, neuronal damage and high antioxidant properties. The aim of this research was to evaluate the neuroprotective, antioxidant and anti-inflammatory activities of luteolin-7-O-glucoside (Lut7). Undifferentiated and retinoic acid (RA)-differentiated SH-SY5Y cells were pretreated with Lut7 and incubated with 6-hydroxydopamine (6-OHDA). Cytotoxic and neuroprotective effects were determined by MTT assay. Antioxidant capacity was determined by DPPH, FRAP, and ORAC assays. ROS production, mitochondrial membrane potential (ΔΨm), Caspase–3 activity, acetylcholinesterase inhibition (AChEI) and nuclear damage were also determined in SH-SY5Y cells. TNF-α, IL-6 and IL-10 release were evaluated in LPS-induced RAW264.7 cells by ELISA. In undifferentiated SH-SY5Y cells, Lut7 increased cell viability after 24 h, while in RA-differentiated SH-SY5Y cells, Lut7 increased cell viability after 24 and 48 h. Lut7 showed a high antioxidant activity when compared with synthetic antioxidants. In undifferentiated cells, Lut7 prevented mitochondrial membrane depolarization induced by 6-OHDA treatment, decreased Caspase-3 and AChE activity, and inhibited nuclear condensation and fragmentation. In LPS-stimulated RAW264.7 cells, Lut7 treatment reduced TNF-α levels and increased IL-10 levels after 3 and 24 h, respectively. In summary, the results suggest that Lut7 has neuroprotective effects, thus, further studies should be considered to validate its pharmacological potential in more complex models, aiming the treatment of neurodegenerative diseases.
Collapse
|
24
|
Shomali N, Suliman Maashi M, Baradaran B, Daei Sorkhabi A, Sarkesh A, Mohammadi H, Hemmatzadeh M, Marofi F, Sandoghchian Shotorbani S, Jarahian M. Dysregulation of Survivin-Targeting microRNAs in Autoimmune Diseases: New Perspectives for Novel Therapies. Front Immunol 2022; 13:839945. [PMID: 35309327 PMCID: PMC8927965 DOI: 10.3389/fimmu.2022.839945] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/14/2022] [Indexed: 12/15/2022] Open
Abstract
It has been well established that the etiopathogenesis of diverse autoimmune diseases is rooted in the autoreactive immune cells' excessively proliferative state and impaired apoptotic machinery. Survivin is an anti-apoptotic and mitotic factor that has sparked a considerable research interest in this field. Survivin overexpression has been shown to contribute significantly to the development of autoimmune diseases via autoreactive immune cell overproliferation and apoptotic dysregulation. Several microRNAs (miRNAs/miRs) have been discovered to be involved in survivin regulation, rendering the survivin-miRNA axis a perspective target for autoimmune disease therapy. In this review, we discuss the role of survivin as an immune regulator and a highly implicated protein in the pathogenesis of autoimmune diseases, the significance of survivin-targeting miRNAs in autoimmunity, and the feasibility of targeting the survivin-miRNA axis as a promising therapeutic option for autoimmune diseases.
Collapse
Affiliation(s)
- Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marwah Suliman Maashi
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Daei Sorkhabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aila Sarkesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Maryam Hemmatzadeh
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siamak Sandoghchian Shotorbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit (G401), Heidelberg, Germany
| |
Collapse
|
25
|
Rivera-Yañez CR, Ruiz-Hurtado PA, Mendoza-Ramos MI, Reyes-Reali J, García-Romo GS, Pozo-Molina G, Reséndiz-Albor AA, Nieto-Yañez O, Méndez-Cruz AR, Méndez-Catalá CF, Rivera-Yañez N. Flavonoids Present in Propolis in the Battle against Photoaging and Psoriasis. Antioxidants (Basel) 2021; 10:antiox10122014. [PMID: 34943117 PMCID: PMC8698766 DOI: 10.3390/antiox10122014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/09/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022] Open
Abstract
The skin is the main external organ. It protects against different types of potentially harmful agents, such as pathogens, or physical factors, such as radiation. Skin disorders are very diverse, and some of them lack adequate and accessible treatment. The photoaging of the skin is a problem of great relevance since it is related to the development of cancer, while psoriasis is a chronic inflammatory disease that causes scaly skin lesions and deterioration of the lifestyle of people affected. These diseases affect the patient's health and quality of life, so alternatives have been sought that improve the treatment for these diseases. This review focuses on describing the properties and benefits of flavonoids from propolis against these diseases. The information collected shows that the antioxidant and anti-inflammatory properties of flavonoids play a crucial role in the control and regulation of the cellular and biochemical alterations caused by these diseases; moreover, flavones, flavonols, flavanones, flavan-3-ols, and isoflavones contained in different worldwide propolis samples are the types of flavonoids usually evaluated in both diseases. Therefore, the research carried out in the area of dermatology with bioactive compounds of different origins is of great relevance to developing preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Claudia Rebeca Rivera-Yañez
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico;
| | - Porfirio Alonso Ruiz-Hurtado
- Laboratorio de Toxicología de Productos Naturales, Departamento de Farmacia, IPN, Escuela Nacional de Ciencias Biológicas, Av. Wilfrido Massieu, Gustavo A. Madero 07738, Mexico;
| | - María Isabel Mendoza-Ramos
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- Laboratorio de Inmunología, Unidad de Morfofisiología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Julia Reyes-Reali
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- Laboratorio de Inmunología, Unidad de Morfofisiología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Gina Stella García-Romo
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- Laboratorio de Inmunología, Unidad de Morfofisiología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Glustein Pozo-Molina
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- Laboratorio de Genética y Oncología Molecular, Laboratorio 5, Edificio A4, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Aldo Arturo Reséndiz-Albor
- Laboratorio de Inmunidad de Mucosas, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Salvador Díaz Mirón y Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, Mexico City 11340, Mexico;
| | - Oscar Nieto-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
| | - Adolfo René Méndez-Cruz
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- Laboratorio de Inmunología, Unidad de Morfofisiología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
| | - Claudia Fabiola Méndez-Catalá
- Laboratorio de Genética y Oncología Molecular, Laboratorio 5, Edificio A4, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- División de Investigación y Posgrado, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Correspondence: (C.F.M.-C.); (N.R.-Y.); Tel.: +52-5522-476-721 (N.R.-Y.)
| | - Nelly Rivera-Yañez
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.I.M.-R.); (J.R.-R.); (G.S.G.-R.); (G.P.-M.); (O.N.-Y.); (A.R.M.-C.)
- División de Investigación y Posgrado, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico
- Correspondence: (C.F.M.-C.); (N.R.-Y.); Tel.: +52-5522-476-721 (N.R.-Y.)
| |
Collapse
|
26
|
Larsen SB, Cowley CJ, Sajjath SM, Barrows D, Yang Y, Carroll TS, Fuchs E. Establishment, maintenance, and recall of inflammatory memory. Cell Stem Cell 2021; 28:1758-1774.e8. [PMID: 34320411 PMCID: PMC8500942 DOI: 10.1016/j.stem.2021.07.001] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/21/2021] [Accepted: 07/01/2021] [Indexed: 01/12/2023]
Abstract
Known for nearly a century but through mechanisms that remain elusive, cells retain a memory of inflammation that equips them to react quickly and broadly to diverse secondary stimuli. Using murine epidermal stem cells as a model, we elucidate how cells establish, maintain, and recall inflammatory memory. Specifically, we landscape and functionally interrogate temporal, dynamic changes to chromatin accessibility, histone modifications, and transcription factor binding that occur during inflammation, post-resolution, and in memory recall following injury. We unearth an essential, unifying role for the general stress-responsive transcription factor FOS, which partners with JUN and cooperates with stimulus-specific STAT3 to establish memory; JUN then remains with other homeostatic factors on memory domains, facilitating rapid FOS re-recruitment and gene re-activation upon diverse secondary challenges. Extending our findings, we offer a comprehensive, potentially universal mechanism behind inflammatory memory and less discriminate recall phenomena with profound implications for tissue fitness in health and disease.
Collapse
Affiliation(s)
- Samantha B. Larsen
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA,New York University School of Medicine, Neuroscience Institute, New York, NY 10016, USA
| | - Christopher J. Cowley
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Sairaj M. Sajjath
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Douglas Barrows
- Bioinformatics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Yihao Yang
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Thomas S. Carroll
- Bioinformatics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA,Lead Contact to whom correspondence should be addressed during the review process:
| |
Collapse
|
27
|
Liu D, Luo H, Qiao C. SHP-1/STAT3 Interaction Is Related to Luteolin-Induced Myocardial Ischemia Protection. Inflammation 2021; 45:88-99. [PMID: 34460026 PMCID: PMC8403691 DOI: 10.1007/s10753-021-01530-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/28/2021] [Indexed: 01/06/2023]
Abstract
Prevention and management of myocardial ischemia/reperfusion (I/R) injury is a key step in coronary heart disease surgery. Luteolin is a falconoid compound that has an antioxidant effect, but its mechanism in I/R injury in vivo and in vitro is still under explored. This study attempted to reveal the role of luteolin (Lut) in I/R through mediation of the Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1)/Signal transducer and activator of transcription 3 (STAT3) pathway. To establish I/R rat models, the left anterior descending artery (LAD) was ligated for 30 min and re-perfused for 1 h in Lut-pretreated or nude rats. Comparisons between infarct area, cardiac dysfunction, and myocardial cell death and inflammatory reaction were performed in I/R-induced rats. Hypoxia/reoxygenation (H/R) cell models were established by stimulating H9c2 cells with 95% nitrogen and 5% carbon dioxide. Simultaneously, H/R-related cell death and inflammatory reactions were investigated following Lut treatment. The target protein of Lut was identified using western blotting. Pro-inflammatory cytokines were also measured in serum or Lut-pretreated cell culture medium. The results revealed that compared with the I/R group, Lut treatment could significantly decrease myocardial infarction (MI) area, increase left ventricular ejection fraction (LVEF), and decrease cell death and pro-inflammatory cytokines in the serum. Decreased apoptosis and inflammatory cytokines were also observed in H/R cells after Lut treatment. Lut treatment downregulated SHP-1 expression and subsequently upregulated STAT3 phosphorylation in both I/R rat heart tissue and H9c2 cells. The findings of the current study suggest that Lut can protect the heart and reduce MI area, cell apoptosis rate, and inflammatory level in I/R models.
Collapse
Affiliation(s)
- Donghai Liu
- Cardiovascular Surgery II, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Hong Luo
- Cardiovascular Surgery II, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chenhui Qiao
- Cardiovascular Surgery II, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China. .,Cardiovascular Surgery II, The First Affiliated Hospital of Zhengzhou University, Henan, 450052, Zhengzhou, China.
| |
Collapse
|
28
|
Alantolactone Suppresses Proliferation and the Inflammatory Response in Human HaCaT Keratinocytes and Ameliorates Imiquimod-Induced Skin Lesions in a Psoriasis-Like Mouse Model. Life (Basel) 2021; 11:life11070616. [PMID: 34202301 PMCID: PMC8303865 DOI: 10.3390/life11070616] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022] Open
Abstract
Psoriasis is an immune-mediated inflammatory disease that affects 2% to 3% of the world population. Alantolactone, a sesquiterpene lactone, was isolated from Inula helenium and Radix inulae and has several biological effects, including antifungal, anthelmintic, antimicrobial, anti-inflammatory, antitrypanosomal, and anticancer properties. This study aimed to evaluate the antipsoriatic potential of alantolactone in vitro and in vivo and to explore its underlying mechanisms. These results showed that alantolactone significantly attenuated IL-17A, IL-22, oncostatin M, IL-1α, and TNF-α (M5) cytokine-induced hyperproliferation in HaCaT keratinocytes. Moreover, M5 cytokines significantly upregulated the mRNA levels of TNF-α, IL-6, IL-1β, and IL-8. However, alantolactone attenuated the upregulation of these inflammatory cytokines. In addition, alantolactone was found to inhibit STAT3 phosphorylation and NF-κB p65 nuclear translocation in HaCaT keratinocytes. Furthermore, alantolactone treatment in mice significantly alleviated the severity of skin lesions (erythema, scaling and epidermal thickness, and inflammatory cell infiltration) and decreased the mRNA expression of inflammatory cytokines (e.g., TNF-α, IL-6, IL-1β, IL-8, IL-17A, and IL-23) in an IMQ-induced-like mouse model. Therefore, our new findings revealed that alantolactone alleviates psoriatic skin lesions by inhibiting inflammation, making it an attractive candidate for future development as an antipsoriatic agent.
Collapse
|
29
|
The Effect of Herbal Medicinal Products on Psoriasis-Like Keratinocytes. Biomolecules 2021; 11:biom11030371. [PMID: 33801280 PMCID: PMC8000521 DOI: 10.3390/biom11030371] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by hyperproliferation of keratinocytes and expression of pro-inflammatory cytokines in the epidermis. New biological drugs were developed for the systemic treatment of moderate to severe psoriasis. However, products for the topical treatment of mild psoriasis are still required. Here, we examined the effect of natural compounds on psoriasis-like keratinocytes in vitro and ex vivo. Psoriasis-like keratinocytes were generated by treating human primary keratinocytes with the psoriasis-associated cytokines IL-17A, TNF-α and IL-22. Initially, 10 botanical extracts from Ayurvedic Medicine, Traditional Chinese Medicine, Northern American traditional medicine and Occidental Monastic Medicine were investigated using BrdU assays and IL-6 and IL-8 ELISAs. Curcuma amada, Humulus lupulus and Hypericum perforatum turned out to be the most effective plant extracts. In vitro, the plant extracts inhibited the expression of anti-microbial peptides (β-defensin 2), the hyperproliferation marker keratin 17, the glucose transporter 1 and downregulated the nuclear translocation of NF-κB and pSTAT3. In an ex vivo psoriasis model, Humulus lupulus displayed the most prominent anti-proliferative and anti-inflammatory effect. In conclusion, among the plant extracts investigated, Humulus lupulus showed the most promising anti-psoriatic effect. It is an interesting candidate for topical psoriasis treatment that should be further studied in clinical trials.
Collapse
|
30
|
Gendrisch F, Esser PR, Schempp CM, Wölfle U. Luteolin as a modulator of skin aging and inflammation. Biofactors 2021; 47:170-180. [PMID: 33368702 DOI: 10.1002/biof.1699] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022]
Abstract
Luteolin belongs to the group of flavonoids and can be found in flowers, herbs, vegetables and spices. It plays an important role in defending plants, for example against UV radiation by partially absorbing UVA and UVB radiation. Thus, luteolin can also decrease adverse photobiological effects in the skin by acting as a first line of defense. Furthermore, anti-oxidative and anti-inflammatory activities of luteolin were described on keratinocytes and fibroblasts as well as on several immune cells (e.g., macrophages, mast cell, neutrophils, dendritic cells and T cells). Luteolin can suppress proinflammatory mediators (e.g., IL-1β, IL-6, IL-8, IL-17, IL-22, TNF-α and COX-2) and regulate various signaling pathway (e.g., the NF-κB, JAK-STAT as well as TLR signaling pathway). In this way, luteolin modulates many inflammatory processes of the skin. The present review summarizes the recent in vitro and in vivo research on luteolin in the field of skin aging and skin cancer, wound healing as well as inflammatory skin diseases, including psoriasis, contact dermatitis and atopic dermatitis. In conclusion, luteolin might be a promising molecule for the development of topic formulations and systemic agents against inflammatory skin diseases.
Collapse
Affiliation(s)
- Fabian Gendrisch
- Research Center Skinitial, Department of Dermatology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Philipp R Esser
- Allergy Research Group, Department of Dermatology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Christoph M Schempp
- Research Center Skinitial, Department of Dermatology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Ute Wölfle
- Research Center Skinitial, Department of Dermatology, Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
31
|
De Stefano A, Caporali S, Di Daniele N, Rovella V, Cardillo C, Schinzari F, Minieri M, Pieri M, Candi E, Bernardini S, Tesauro M, Terrinoni A. Anti-Inflammatory and Proliferative Properties of Luteolin-7-O-Glucoside. Int J Mol Sci 2021; 22:1321. [PMID: 33525692 PMCID: PMC7865871 DOI: 10.3390/ijms22031321] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Flavonoids display a broad range of structures and are responsible for the major organoleptic characteristics of plant-derived foods and beverages. Recent data showed their activity, and in particular of luteolin-7-O-glucoside (LUT-7G), in reduction of oxidative stress and inflammatory mechanisms in different physiological systems. In this paper, we tried to elucidate how LUT-7G could exert both antioxidant and anti-inflammatory effects in endothelial cells cultured in vitro. Here, we showed that LUT-7G is able to inhibit the STAT3 pathway, to have an antiproliferative action, and an important antioxidant property in HUVEC cells. These properties are exerted by the flavone in endothelial through the transcriptional repression of a number of inflammatory cytokines and their receptors, and by the inhibition of ROS generation. ROS and STAT3 activation has been correlated with the production of oxysterols and other hydroxylated fatty acids, and they have been recognized important as players of atherogenesis and cardiocirculatory system diseases. The analysis of the general production pathway of these hydroxylated species, showed a strong decrease of cholesterol hydroxylated species such as 7-alpha-hydroxicholesterol, 7-beta-hydroxicholesterol by the treatment with LUT-7G. This confirms the anti-inflammatory properties of LUT-7G also in the endothelial district, showing for the first time the molecular pathway that verify previous postulated cardiovascular benefits of this flavone.
Collapse
Affiliation(s)
- Alessandro De Stefano
- Centre of Space Biomedicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.D.S.); (N.D.D.); (V.R.); (M.T.)
| | - Sabrina Caporali
- Department of Industrial Engineering, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Nicola Di Daniele
- Centre of Space Biomedicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.D.S.); (N.D.D.); (V.R.); (M.T.)
| | - Valentina Rovella
- Centre of Space Biomedicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.D.S.); (N.D.D.); (V.R.); (M.T.)
| | - Carmine Cardillo
- Department of Clinical Sciences and Translational Medicine, Cattolica University of Rome, Via Montpellier, 1, 00133 Rome, Italy;
- Internal Medicine, Policlinico A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, 00133 Rome, Italy;
| | - Francesca Schinzari
- Internal Medicine, Policlinico A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, 00133 Rome, Italy;
| | - Marilena Minieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy; (M.M.); (M.P.); (E.C.); (S.B.)
| | - Massimo Pieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy; (M.M.); (M.P.); (E.C.); (S.B.)
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy; (M.M.); (M.P.); (E.C.); (S.B.)
- Laboratory of Biochemistry, IDI-IRCCS Fondazione Luigi Maria Monti, Via Monti di Creta 104, 00167 Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy; (M.M.); (M.P.); (E.C.); (S.B.)
| | - Manfredi Tesauro
- Centre of Space Biomedicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (A.D.S.); (N.D.D.); (V.R.); (M.T.)
| | - Alessandro Terrinoni
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy; (M.M.); (M.P.); (E.C.); (S.B.)
| |
Collapse
|
32
|
Wen K, Fang X, Yang J, Yao Y, Nandakumar KS, Salem ML, Cheng K. Recent Research on Flavonoids and their Biomedical Applications. Curr Med Chem 2021; 28:1042-1066. [PMID: 32660393 DOI: 10.2174/0929867327666200713184138] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 02/14/2020] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
Flavonoids, commonly found in various plants, are a class of polyphenolic compounds having a basic structural unit of 2-phenylchromone. Flavonoid compounds have attracted much attention due to their wide biological applications. In order to facilitate further research on the biomedical application of flavonoids, we surveyed the literature published on the use of flavonoids in medicine during the past decade, documented the commonly found structures in natural flavonoids, and summarized their pharmacological activities as well as associated mechanisms of action against a variety of health disorders including chronic inflammation, cancer, cardiovascular complications and hypoglycemia. In this mini-review, we provide suggestions for further research on the biomedical applications of flavonoids.
Collapse
Affiliation(s)
- Kangmei Wen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaochuan Fang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Junli Yang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yongfang Yao
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | | | | | - Kui Cheng
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
33
|
Antioxidant Efficacy of Olive By-Product Extracts in Human Colon HCT8 Cells. Foods 2020; 10:foods10010011. [PMID: 33374501 PMCID: PMC7822158 DOI: 10.3390/foods10010011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/16/2020] [Accepted: 12/19/2020] [Indexed: 12/11/2022] Open
Abstract
The production of olive oil is accompanied by the generation of a huge amount of waste and by-products including olive leaves, pomace, and wastewater. The latter represents a relevant environmental issue because they contain certain phytotoxic compounds that may need specific treatments before the expensive disposal. Therefore, reducing waste biomass and valorizing by-products would make olive oil production more sustainable. Here, we explore the biological actions of extracts deriving from olive by-products including olive pomace (OP), olive wastewater (OWW), and olive leaf (OLs) in human colorectal carcinoma HCT8 cells. Interestingly, with the same phenolic concentration, the extract obtained from the OWW showed higher antioxidant ability compared with the extracts derived from OP and OLs. These biological effects may be related to the differential phenolic composition of the extracts, as OWW extract contains the highest amount of hydroxytyrosol and tyrosol that are potent antioxidant compounds. Furthermore, OP extract that contains a higher level of vanillic acid than the other extracts displayed a cytotoxic action at the highest concentration. Together these findings revealed that phenols in the by-product extracts may interfere with signaling molecules that cross-link several intracellular pathways, raising the possibility to use them for beneficial health effects.
Collapse
|
34
|
Shao L, Xiong X, Zhang Y, Miao H, Ren Y, Tang X, Song J, Wang C. IL-22 ameliorates LPS-induced acute liver injury by autophagy activation through ATF4-ATG7 signaling. Cell Death Dis 2020; 11:970. [PMID: 33177520 PMCID: PMC7658242 DOI: 10.1038/s41419-020-03176-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022]
Abstract
Uncontrollable inflammatory response acts as a driver of sepsis-associated liver injury (SALI). IL-22 plays an important role in regulating inflammatory responses, but its role in SALI remains unknown. The aim of the study was to assess the association of serum IL-22 with SALI in pediatric patients and to enclose the underlying mechanisms of IL-22 involved in lipopolysaccharide (LPS) - induced acute liver injury (ALI) in mice. Serum IL-22 levels in patients with SALI were significantly lower than in septic patients without liver injury, and the area under receiver operating characteristic (ROC) curve of IL-22 for discriminating SALI was 0.765 (95% CI: 0.593-0.937). Pre-administration of recombinant murine IL-22 alleviated LPS-induced ALI in mice, and serum IL-6 levels and the mRNA levels of TNF-α, IL-1β, and IL-6 in livers were decreased in response to IL-22 pre-treatment in mice. More importantly, IL-22 pre-treatment activated hepatic autophagy mediated by activating transcription factor 4 (ATF4)-autophagy-related gene 7 (ATG7) signaling in vivo and in vitro in response to LPS administration. Moreover, knockdown of ATF4 in mice aggravated LPS-induced ALI, which was associated with suppressed ATG7-related autophagy. In addition, the protective effects of IL-22 on LPS-induced ALI was partially blocked by ATF4 knockdown, which was associated with lower expression of LC3II/I in the livers of ATF4 knockdown (HT or Atf4+/-) mice compared with wild-type mice (WT or Atf4+/+) mice. In conclusion, low serum IL-22 level is associated with SALI occurrence, and IL-22 pre-administration activates autophagy in hepatocytes and protects mice against LPS-induced ALI partially related to ATF4-ATG7 signaling pathway.
Collapse
Affiliation(s)
- Lujing Shao
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Xi Xiong
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Yucai Zhang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China.
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062, China.
| | - Huijie Miao
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Yuqian Ren
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Xiaomeng Tang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Jia Song
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Chunxia Wang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China.
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062, China.
| |
Collapse
|
35
|
Wang W, Wang H, Zhao Z, Huang X, Xiong H, Mei Z. Thymol activates TRPM8-mediated Ca 2+ influx for its antipruritic effects and alleviates inflammatory response in Imiquimod-induced mice. Toxicol Appl Pharmacol 2020; 407:115247. [PMID: 32971067 DOI: 10.1016/j.taap.2020.115247] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/06/2020] [Accepted: 09/18/2020] [Indexed: 12/24/2022]
Abstract
Psoriasis is a highly prevalent chronic dermatitis, characterized by widespread skin inflammation and spontaneous itch. Given the adverse reactions and drug dependence of current treatment, new drugs for psoriasis therapy are urgently needed. This study aims to explore the anti-psoriatic effects of thymol in imiquimod (IMQ) induced mice, and elucidate the potential mechanisms for its therapeutic activities. Thymol reduced the scratching behavior in IMQ mice, and activated Ca2+ response in cervical DRG neurons via TRPM8 channel. Also, thymol alleviated psoriasis-like skin lesions, and attenuated the enhanced infiltration of dermal neutrophils, dendritic cells (DCs) and Th17 cells. In addition, it reversed the upregulated expression of pro-inflammatory cytokines in the skin (TNF-α, IL-22, IL-23, IL-17A, IL-17F, IL-17C, IL-6, IL-1β and IFN-γ) and serum (TNF-α, IL-6, IL-1β, IL-17A and IFN-γ). Our results indicated that thymol can effectively ameliorate pruritus and the symptoms of psoriasis-like inflammation induced by IMQ, which makes it a promising drug for the treatment of psoriasis.
Collapse
Affiliation(s)
- Wen Wang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Hua Wang
- College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Zhongqiu Zhao
- Washington University School of Medicine, St. Louis, MO 63110, United States; Barnes-Jewish Hospital, St. Louis, MO 63110, United States
| | - Xiaoqing Huang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education), School of Pharmaceutical Sciences, Wuhan University, Wuhan 430072, China
| | - Hairong Xiong
- College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China.
| | - Zhinan Mei
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China.
| |
Collapse
|
36
|
Chen H, Liu H, Tang B, Chen Y, Han L, Yu J, Yan Y, Lu C. The Protective Effects of 18 β-Glycyrrhetinic Acid on Imiquimod-Induced Psoriasis in Mice via Suppression of mTOR/STAT3 Signaling. J Immunol Res 2020; 2020:1980456. [PMID: 32908937 PMCID: PMC7474397 DOI: 10.1155/2020/1980456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/09/2020] [Indexed: 02/08/2023] Open
Abstract
Psoriasis is recognized as an autoimmune and inflammatory dermatosis, which is estimated to affect 2-3% of the population worldwide. 18β-Glycyrrhetinic acid (GA), one of the main ingredients of Licorice (Glycyrrhiza glabra L.), has been shown to have numerous pharmacological effects such as antioxidative, antitumor, and anti-inflammatory activities. However, it remains to be explored whether GA has antipsoriatic effect on psoriasis. In this study, we evaluated the protective effect of GA on psoriasis and its mechanisms of action in imiquimod-induced psoriasis-like mouse model. Results indicated that GA dramatically improved psoriatic lesions and reduced psoriasis area and severity index scores. GA also suppressed the mRNA levels of IL-6, TNF-α, IL-17, IL-23, and IL-1β in the skin and increased the proportion of CD4+ Foxp3+ regulatory T cells (Tregs) in both lymph nodes and spleens. Its anti-inflammatory and immunomodulatory activities may be related to its suppression of the STAT3 and mTOR signaling. In conclusion, GA ameliorated the symptoms of psoriasis, at least in part, through inhibition of inflammatory cytokines and STAT3/mTOR signaling and activation of Tregs in both lymph nodes and spleens. These effects are expected to be beneficial in the treatment and prevention of psoriasis.
Collapse
Affiliation(s)
- Haiming Chen
- State Key laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510115, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510115, China
| | - Huazhen Liu
- State Key laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510115, China
| | - Bin Tang
- State Key laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510115, China
| | - Yuchao Chen
- State Key laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510115, China
| | - Ling Han
- State Key laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510115, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510115, China
| | - Jingjie Yu
- State Key laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510115, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510115, China
| | - Yuhong Yan
- State Key laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510115, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510115, China
| | - Chuanjian Lu
- State Key laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510115, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510115, China
| |
Collapse
|
37
|
Furue M. Regulation of Filaggrin, Loricrin, and Involucrin by IL-4, IL-13, IL-17A, IL-22, AHR, and NRF2: Pathogenic Implications in Atopic Dermatitis. Int J Mol Sci 2020; 21:E5382. [PMID: 32751111 PMCID: PMC7432778 DOI: 10.3390/ijms21155382] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 12/16/2022] Open
Abstract
Atopic dermatitis (AD) is an eczematous, pruritic skin disorder with extensive barrier dysfunction and elevated interleukin (IL)-4 and IL-13 signatures. The barrier dysfunction correlates with the downregulation of barrier-related molecules such as filaggrin (FLG), loricrin (LOR), and involucrin (IVL). IL-4 and IL-13 potently inhibit the expression of these molecules by activating signal transducer and activator of transcription (STAT)6 and STAT3. In addition to IL-4 and IL-13, IL-22 and IL-17A are probably involved in the barrier dysfunction by inhibiting the expression of these barrier-related molecules. In contrast, natural or medicinal ligands for aryl hydrocarbon receptor (AHR) are potent upregulators of FLG, LOR, and IVL expression. As IL-4, IL-13, IL-22, and IL-17A are all capable of inducing oxidative stress, antioxidative AHR agonists such as coal tar, glyteer, and tapinarof exert particular therapeutic efficacy for AD. These antioxidative AHR ligands are known to activate an antioxidative transcription factor, nuclear factor E2-related factor 2 (NRF2). This article focuses on the mechanisms by which FLG, LOR, and IVL expression is regulated by IL-4, IL-13, IL-22, and IL-17A. The author also summarizes how AHR and NRF2 dual activators exert their beneficial effects in the treatment of AD.
Collapse
Affiliation(s)
- Masutaka Furue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka 812-8582, Japan; ; Tel.: +81-92-642-5581; Fax: +81-92-642-5600
- Research and Clinical Center for Yusho and Dioxin, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka 812-8582, Japan
- Division of Skin Surface Sensing, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka 812-8582, Japan
| |
Collapse
|
38
|
Wang J, Kaplan N, Wang S, Yang W, Wang L, He C, Peng H. Autophagy plays a positive role in induction of epidermal proliferation. FASEB J 2020; 34:10657-10667. [PMID: 32598088 DOI: 10.1096/fj.202000770rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/12/2020] [Accepted: 05/29/2020] [Indexed: 12/09/2022]
Abstract
Autophagy is a multistage catabolic process that mediates stress responses. However, the role of autophagy in epidermal proliferation, particularly under conditions when the epidermis becomes "activated" (hyperproliferative), remains unclear. We have shown that inhibition of Beclin 1, a key activator in the initiation phase of autophagy, attenuates imiquimod (IMQ)-induced epidermal hyperplasia in adult mice as well as naturally occurring hyperproliferation in neonatal mouse epidermis. Inhibition of Beclin 1 did not change the levels of several key inflammatory molecules or the numbers of immune cells in lesional skins. This indicates that autophagy does not affect inflammatory regulators in IMQ-treated mouse skin. Bioinformatic analysis combined with gene expression quantitative assays, revealed that a deficiency in autophagy decreases the expression of PDZ Binding Kinase (PBK), a regulator of the cell cycle, in mouse epidermis and human epidermal keratinocytes (HEKs). Interestingly, the decrease in PBK results in inhibition of proliferation in HEKs and such reduced proliferation can be rescued by activation of p38, the downstream signaling of PBK. Collectively, autophagy plays a positive role in epidermal proliferation, which is in part via regulating PBK expression.
Collapse
Affiliation(s)
- Junyi Wang
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States.,Department of Ophthalmology, The First Center of the PLA General Hospital, Beijing, China
| | - Nihal Kaplan
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Sijia Wang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wending Yang
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Liqiang Wang
- Department of Ophthalmology, The First Center of the PLA General Hospital, Beijing, China
| | - Congcong He
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Han Peng
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| |
Collapse
|
39
|
Asgharian P, Delazar A, Asnaashari S. Chemical Constituents of Eremostachys macrophylla Montbr. & Auch. Aerial Parts. PHARMACEUTICAL SCIENCES 2020. [DOI: 10.34172/ps.2019.65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background:
Eremostachys macrophylla Montbr. & Auch. is one of the wild growing species of herbs found in East Azerbaijan province of Iran. These species are used in folk medicine for the healing of wound, treatment of snake bites, rheumatism and joint pains. The primary aim of this study was to obtain natural pure compounds and this was done by subjecting the aerial parts of Eremostachys macrophylla Montbr. & Auch. to phytochemical analysis. Methods: The air-dried and crushed aerial parts were respectively extracted with n-hexane, dichloromethane (DCM) and methanol (MeOH) solvents using a soxhlet apparatus. The 10%, 20% and 40% of MeOH in water Sep-Pak fractions of the MeOH extract were subjected to a preparative reversed- phase high performance liquid chromatography (RP-HPLC). Also, the isolated pure compounds were identified by one-dimensional nuclear magnetic resonance (1D-NMR) spectroscopic technique. Results: The results obtained in this study showed the presence of seven pure components; (1) Lamalbide, (2) Sesamoside, (3) Phlomiol, (4) Verbascoside, (5) Luteolin-7-O- glucoside, (6) Apigenin-7-O- rutinoside and (7) Kaempferol-3-O- glucoside with iridoid, phenylethanoid and flavonoid structures. Conclusion: The results from the study demonstrated that the aerial parts of E. macrophylla could be a good source of iridoids, phenylethanoids and flavonoids.
Collapse
Affiliation(s)
- Parina Asgharian
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Delazar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Asnaashari
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
40
|
Mercurio L, Failla CM, Capriotti L, Scarponi C, Facchiano F, Morelli M, Rossi S, Pagnanelli G, Albanesi C, Cavani A, Madonna S. Interleukin (IL)-17/IL-36 axis participates to the crosstalk between endothelial cells and keratinocytes during inflammatory skin responses. PLoS One 2020; 15:e0222969. [PMID: 32352958 PMCID: PMC7192413 DOI: 10.1371/journal.pone.0222969] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 04/15/2020] [Indexed: 12/22/2022] Open
Abstract
In inflammatory skin conditions, such as psoriasis, vascular enlargement is associated with endothelial cell proliferation, release of cytokines and adhesion molecule expression. Interleukin (IL)-17A is a pro-inflammatory cytokine mainly secreted by T helper-17 cells that is critically involved in psoriasis pathogenesis. IL-36α, IL-36β and IL-36γ are also inflammatory cytokines up-regulated in psoriasis and induced by various stimuli, including IL-17A. In this study, we found that human keratinocytes are the main source of IL-36, in particular of IL-36γ. This cytokine was strongly induced by IL-17A and, together with IL-17A, efficiently activated human dermal microvascular endothelial cells (HDMECs), which expressed both IL-17 and IL-36 receptors. Both IL-36γ and IL-17A induced cell proliferation through specific molecular cascades involving ERK1/2 only or ERK1/2, STAT3 and NF-κB, respectively. We highlighted the intense IL-17A- and IL-36γ -dependent interplay between keratinocytes and HDMECs, likely active in the psoriatic lesions and leading to the establishment of a cytokine network responsible for the development and maintenance of the inflamed state. IL-17A or IL-36γ showed in HDMECs a synergic activity with TNF-α by potently inducing inflammatory cytokine/chemokine release and ICAM-1 expression. We also investigated the involvement of IL-36γ and VEGF-A, substantially reduced in lesional skin of psoriatic patients pharmacologically treated with the anti-IL-17A antibody Secukinumab. Importantly, keratinocyte-derived IL-36γ represented an additional pro-angiogenic mediator of IL-17A. We observed that keratinocyte-derived VEGF-A influenced proliferation but did not act on expression of adhesion molecules in HDMECs. On the other hand, inhibition of IL-36γ released by IL-17A-treated keratinocytes impaired either proliferation or ICAM-1 expression both in HDMECs and in an in vivo murine model of psoriasis. Taken together, our data demonstrated that IL-17A and IL-36γ are highly involved in endothelial cells/keratinocytes crosstalk in inflammatory skin conditions.
Collapse
Affiliation(s)
- Laura Mercurio
- Laboratory of Experimental Immunology, IDI-IRCCS, Rome, Italy
| | | | | | | | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Martina Morelli
- Laboratory of Experimental Immunology, IDI-IRCCS, Rome, Italy
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (ISS), Rome, Italy
| | | | | | - Andrea Cavani
- National Institute for Health, Migration and Poverty (NIHMP), Rome, Italy
| | | |
Collapse
|
41
|
Luteolin retards CXCL12-induced Jurkat cells migration by disrupting transcription of CXCR4. Exp Mol Pathol 2020; 113:104370. [PMID: 31917965 DOI: 10.1016/j.yexmp.2020.104370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 12/23/2019] [Accepted: 01/04/2020] [Indexed: 12/13/2022]
|
42
|
Diluvio L, Caporali S, Lozzi F, Campione E, Mazzilli S, Lanna C, Bianchi L, Bernardini S, Minieri M, Mauriello A, Ferlosio A, Candi E, Terrinoni A. Birt-Hogg-Dubé syndrome, from non-invasive dermatologic assessment to gene testing, molecular and ultrastructural histologic analysis. J Eur Acad Dermatol Venereol 2020; 34:e206-e209. [PMID: 31876332 DOI: 10.1111/jdv.16168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- L Diluvio
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - S Caporali
- Department of Industrial Engineering, University of Rome Tor Vergata, Rome, Italy
| | - F Lozzi
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - E Campione
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - S Mazzilli
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - C Lanna
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - L Bianchi
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - S Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - M Minieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - A Mauriello
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - A Ferlosio
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - E Candi
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy.,Biochemistry Laboratory, Fondazione Luigi Maria Monti, IDI-IRCCS, Rome, Italy
| | - A Terrinoni
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
43
|
Miao X, Xiang Y, Mao W, Chen Y, Li Q, Fan B. TRIM27 promotes IL-6-induced proliferation and inflammation factor production by activating STAT3 signaling in HaCaT cells. Am J Physiol Cell Physiol 2019; 318:C272-C281. [PMID: 31747314 DOI: 10.1152/ajpcell.00314.2019] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The IL-6/STAT3 signaling pathway is required for the development of psoriatic lesions, and tripartite motif-containing 27 (TRIM27) is a protein inhibitor of activated STAT3 (PIAS3)-interacting protein that could modulate IL-6-induced STAT3 activation. However, whether TRIM27 is associated with the IL-6/STAT3 signaling pathway in psoriasis remains enigmatic. TRIM27 expression and gene set enrichment analysis in patients with psoriasis were determined using bioinformatics. Human keratinocyte HaCaT cells treated with recombinant protein IL-6 (rh-IL-6) were transduced with lentivirus silencing TRIM27 and/or PIAS3 or, otherwise, transduced with lentivirus expressing TRIM27 and/or lentivirus silencing STAT3, or MG132, a proteasome-specific protease inhibitor. Cell proliferation and inflammation factor production were measured using Cell Counting Kit-8 and ELISA, respectively. TRIM27, proliferation marker protein Ki-67 (Ki67), phospho-STAT3 (p-STAT3), STAT3, and PIAS3 expressions were determined using real-time quantitative PCR, immunofluorescence staining, or Western blot analysis. Coimmunoprecipitation combined with ubiquitination analysis was performed to explore the interaction between TRIM27 and PIAS3. In the present study, TRIM27 expression was increased in psoriatic lesions, associated with the IL-6 signaling pathway, and induced by rh-IL-6 in a time-dependent manner. The increased cell proliferation, inflammation factor production, and expression of Ki67 and of p-STAT3 relative to STAT3 induced by rh-IL-6 and TRIM27 overexpression were significantly inhibited by TRIM27 silencing and STAT3 silencing, respectively. More importantly, TRIM27 interacted with PIAS3, and its overexpression promoted PIAS3 ubiquitination in HaCaT cells. PIAS3 silencing also significantly promoted TRIM27-dependent and IL6-induced STAT3 activation, cell proliferation, and inflammation factor production. In conclusion, our results highlight that TRIM27 expression is significantly increased by IL-6 and suggest a TRIM27/STAT3-dependent mechanism for regulation of inflammation and proliferation-associated development of psoriasis.
Collapse
Affiliation(s)
- Xiao Miao
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanwei Xiang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiwei Mao
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiran Chen
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bin Fan
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
44
|
Ding X, Zheng L, Yang B, Wang X, Ying Y. Luteolin Attenuates Atherosclerosis Via Modulating Signal Transducer And Activator Of Transcription 3-Mediated Inflammatory Response. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3899-3911. [PMID: 31819365 PMCID: PMC6874161 DOI: 10.2147/dddt.s207185] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022]
Abstract
Background Inflammatory factors play a crucial role throughout the development and progression of atherosclerosis, which has been considered as a chronic vascular inflammatory disease. Luteolin, a natural flavonoid which exists in many natural medicinal materials, has anti-inflammatory, anti-fibrotic and other pharmacological effects. Recently, the protective effects of luteolin on the cardiovascular disease have been reported. However, there is a paucity of studies on anti-atherosclerosis. Therefore, the anti-atherosclerosis potential of luteolin remains to be elucidated. Method ApoE-/- mice were fed with a high-fat diet to induce atherosclerosis in an animal model, where they were treated with oral administration of luteolin for 12 weeks. Primary mouse peritoneal macrophages challenged with oxidized low-density lipoprotein (oxLDL) were used for in vitro mechanistic study. The effectiveness of luteolin in the ApoE-/- mouse model of atherosclerosis was estimated in the aortic sinus and enface, and the underlying mechanisms were explored by molecular modeling study and siRNA-induced gene silencing. Results Our results showed that luteolin remarkably attenuated atherosclerosis in high-fat diet-induced ApoE-/- mouse via alleviating inflammation. We further found that luteolin decreased oxLDL-induced inflammation by inhibiting signal transducer and activator of transcription 3 (STAT3) in vitro, respectively. Further molecular modeling analysis indicated that luteolin interacted with STAT3 primarily through hydrogen bond interaction. Conclusion Luteolin could be a promising candidate molecule for atherosclerosis, and STAT3 may be a potential therapeutic target that could prevent the development of atherosclerosis.
Collapse
Affiliation(s)
- Xiaoji Ding
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Lulu Zheng
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Bo Yang
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Xiaodong Wang
- Department of Vascular Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Yin Ying
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| |
Collapse
|
45
|
Qin L, Chen Z, Yang L, Shi H, Wu H, Zhang B, Zhang W, Xu Q, Huang F, Wu X. Luteolin-7-O-glucoside protects dopaminergic neurons by activating estrogen-receptor-mediated signaling pathway in MPTP-induced mice. Toxicology 2019; 426:152256. [PMID: 31381935 DOI: 10.1016/j.tox.2019.152256] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 06/23/2019] [Accepted: 07/29/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder that is characterized by the degeneration of dopaminergic neurons in substantia nigra (SN). Accumulating evidences implicate the beneficial role of estrogen in the therapy of PD. METHODS In the present study, the protective function of luteolin-7-O-glucoside (LUT-7G), a natural flavonoid, was investigated in 1-methyl-4-phenylpyridinium (MPP+) treated SH-SY5Y cells and 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) induced mice. RESULTS Pre-treatment of LUT-7G increased the viability and reduced the apoptosis of SH-SY5Y cells treated by MPP+. At molecular level, the Bcl-2/Bax ratio was increased, while the expression of cleaved caspase 3 was markedly lessened. Moreover, LUT-7G increased the expression of estrogen receptor (ER), ERα and ERβ, and enhanced the activation of ERK1/2/STAT3/c-Fos that could be abolished by ER antagonists. Furthermore, in vivo experiment indicated that pre-treatment of LUT-7G improved the bradykinesia, and enhanced the muscle strength as well as the balancing capacity of mice treated with MPTP. And LUT-7G prevented the injury of TH positive cells in substantia nigra and increased TH positive nerve fibers in striatum. In addition, pre-treatment of LUT-7G also significantly diminished the MPTP-induced gliosis in substantia nigra. CONCLUSIONS LUT-7G effectively protected dopaminergic neurons against MPP+ or MPTP-induced toxicity, probably by activating the ER-mediated signaling pathway. Our findings explore the therapeutic potential of LUT-7G for PD therapy.
Collapse
Affiliation(s)
- Liyue Qin
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziyu Chen
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu Yang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hailian Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Beibei Zhang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiqi Zhang
- Laboratory of Molecular Psychiatry, Department of Psychiatry, University of Münster, Münster, Germany
| | - Qi Xu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fei Huang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
46
|
Nutritional constituents, health benefits and processing of Rosa Roxburghii: A review. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103456] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
47
|
Michaletti A, Mancini M, Smirnov A, Candi E, Melino G, Zolla L. Multi-omics profiling of calcium-induced human keratinocytes differentiation reveals modulation of unfolded protein response signaling pathways. Cell Cycle 2019; 18:2124-2140. [PMID: 31291818 DOI: 10.1080/15384101.2019.1642066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
By proteomic, metabolomic and transcriptomic approaches we shed light on the molecular mechanism by which human keratinocytes undergo to terminal differentiation upon in vitro calcium treatment. Proteomic analysis revealed a selective induction of the ribosomal proteins RSSA, an inhibitor of cell proliferation and inducer of differentiation, HSP 60, a protein folding chaperone and GRP78, an unfolding protein response signal. Additionally, we observed an induction of EF1D, a transcription factor for genes that contain heat-shock responsive elements. Conversely, RAD23, a protein involved in regulating ER-associated protein degradation was down-regulated. All these modifications indicated an ER stress response, which in turn activated the unfolded protein response signaling pathway through ATF4, as confirmed both by the modulation of amino acids metabolism genes, such as XBP1, PDI and GPR78, and by the metabolomic analysis. Finally, we detected a reduction of PDI protein, as confirmed by the increase of oxidized glutathione. Metabolome analysis indicated that glycolysis failed to fuel the Krebs cycle, which continued to decrease during differentiation, at glance with the PPP pathway, allowing NADH production and glutathione reduction. Since unfolded protein response is linked to keratinization, these results may be useful for studying pathological mechanisms as well as potential treatments for different pathological conditions. Abbreviation: UPR, unfolded protein response; HEK, human epidermal keratinocytes; HKGS, human keratinocytes growth factor.
Collapse
Affiliation(s)
- Anna Michaletti
- a Department of Ecological and Biological Sciences (DEB), University of Tuscia , Viterbo , Italy
| | - Mara Mancini
- b Biochemistry Laboratory, Istituto Dermopatico dell'Immacolata, IDI-IRCCS , Rome , Italy
| | - Artem Smirnov
- c Department of Experimental Medicine, University of Rome "Tor Vergata" , Rome , Italy
| | - Eleonora Candi
- b Biochemistry Laboratory, Istituto Dermopatico dell'Immacolata, IDI-IRCCS , Rome , Italy.,c Department of Experimental Medicine, University of Rome "Tor Vergata" , Rome , Italy
| | - Gerry Melino
- c Department of Experimental Medicine, University of Rome "Tor Vergata" , Rome , Italy.,d MRC Toxicology Unit, Cambridge University , Leicester , UK
| | - Lello Zolla
- e Agriculture and Forest Sciences (DAFNE), University of Tuscia , Viterbo , Italy
| |
Collapse
|
48
|
Luteolin-7-glucoside Promotes Human Epidermal Stem Cell Proliferation by Upregulating β-Catenin, c-Myc, and Cyclin Expression. Stem Cells Int 2019; 2019:1575480. [PMID: 31281367 PMCID: PMC6589269 DOI: 10.1155/2019/1575480] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/31/2019] [Accepted: 04/21/2019] [Indexed: 02/02/2023] Open
Abstract
Skin epidermal stem cells (EpSCs) play critical roles in skin homeostasis and the repair of skin injury. Luteolin-7-glucoside (L7G) has been reported to accelerate skin wound healing through its anti-inflammatory and antioxidative activity. But its effect on EpSCs is not clear. In the present study, we examined the effect of L7G on the proliferation of human EpSCs and explored the mechanisms involved. MTT assay showed that L7G promoted EpSC proliferation in a dose- and time-dependent manner. BrdU incorporation assay and Ki67 immunofluorescence staining confirmed the proproliferative effect of L7G on EpSCs. Cell cycle analysis showed that treatment of EpSCs with L7G decreased the cell number in the G1 phase and increased the cell number in the S phase. In addition, L7G significantly enhanced EpSC migration. Mechanistic studies showed that L7G significantly induced the expression of β-catenin and c-Myc, as well as cyclins D1, A2, and E1 which are critical for G1/S phase transition. L7G stimulated EpSC proliferation through β-catenin and c-Myc. We further examined the effect of L7G on EpSC proliferation in skin tissues by treatment of human skin explants with L7G and examined the number of EpSCs by immunohistochemical stain of EpSC markers α 6 integrin and β 1 integrin. We found that treatment of human skin tissue explants with L7G significantly increased the thickness of the epidermis and increased the numbers of α 6 integrin-positive and β 1 integrin-positive cells at the basal layer of the epidermis. Taken together, these results indicate that L7G promotes EpSC proliferation through upregulating β-catenin, c-Myc, and cyclin expression. L7G can be used to expand EpSCs for generating epidermal autografts and engineered skin equivalents.
Collapse
|
49
|
Palombo R, Caporali S, Falconi M, Iacovelli F, Morozzo Della Rocca B, Lo Surdo A, Campione E, Candi E, Melino G, Bernardini S, Terrinoni A. Luteolin-7- O-β-d-Glucoside Inhibits Cellular Energy Production Interacting with HEK2 in Keratinocytes. Int J Mol Sci 2019; 20:ijms20112689. [PMID: 31159225 PMCID: PMC6600217 DOI: 10.3390/ijms20112689] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/06/2019] [Accepted: 05/15/2019] [Indexed: 01/10/2023] Open
Abstract
Flavonoids have been demonstrated to affect the activity of many mammalian enzyme systems. Their functional phenolic groups are able to mediate antioxidant effects by scavenging free radicals. Molecules of this class have been found able to modulate the activity of kinases, phospholipase A2, cyclooxygenases, lipoxygenase, glutathione S-transferase, and many others. Recently, it has been demonstrated that luteolin, in the form of Luteolin-7-O-β-d-glucoside (LUT-7G) is able to induce the keratinocyte differentiation process in vitro. This flavonoid is able to counteract the proliferative effects of IL-22/IL6 pathway by the inhibition of STAT3 activity also in vivo in a psoriatic mouse model. Observations on energy metabolism changes of differentiating cells led us to perform a complete metabolomics analysis using human primary keratinocytes treated with LUT-7G. Our results show that LUT-7G, is not only able to impair the nuclear translocation of STAT3, but it also blocks the energy metabolism pathway, depressing the glycolytic and Krebs pathway by the inhibition of hexokinase 2 activity. These data confirm that LUT-7G can be proposed as a potential candidate for the treatment of inflammatory and proliferative diseases, but its role as a hexokinase 2 (HEK2) inhibitor opens new perspectives in nutritional science, and especially in cancer therapy, in which the inhibition of the Warburg effect could be relevant.
Collapse
Affiliation(s)
- Ramona Palombo
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
- Laboratory of Cellular and Molecular Neurobiology, Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy.
| | - Sabrina Caporali
- Department of Industrial Engineering, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Mattia Falconi
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy.
| | - Federico Iacovelli
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy.
| | - Blasco Morozzo Della Rocca
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy.
| | - Alessandro Lo Surdo
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| | - Elena Campione
- Department of Systems Medicine, Dermatologic Unit, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
- IDI-IRCCS, Biochemistry Laboratory, via dei Monti di Creta, 104, 00167 Rome, Italy.
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| | - Alessandro Terrinoni
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| |
Collapse
|
50
|
Zeng F, Liu H, Lu D, Liu Q, Chen H, Zheng F. Integrated analysis of gene expression profiles identifies transcription factors potentially involved in psoriasis pathogenesis. J Cell Biochem 2019; 120:12582-12594. [PMID: 30825251 DOI: 10.1002/jcb.28525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 01/03/2019] [Accepted: 02/07/2019] [Indexed: 02/04/2023]
Abstract
Psoriasis is a common inflammatory skin disease mediated by cells and molecules in both the innate and adaptive immune systems. Recently, gene expression profile analysis revealed a large set of immune-related differentially expressed genes (DEGs) in psoriasis. However, the associations between these DEGs and their transcriptional regulation mechanisms have not been completely elucidated. In this study, several psoriasis Gene Expression Omnibus data sets were systematically analyzed using bioinformatics tools to uncover important transcription factors (TFs) that regulate the expression of immune-related DEGs and further enhance our understanding of psoriasis pathogenesis. Common DEGs encoding chemokines, cytokines, antimicrobial peptides, and keratins were identified in psoriasis, and extensive correlations existed among these DEGs. Several common TFs that bind the promoters of the DEGs, including the well-known signal transducer and activator of transcription 1 (STAT1), STAT3, and nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) as well as ETS homologous factor (EHF), Fos-like antigen 1 (FOSL1), and Forkhead box C1 (FOXC1), which are rarely studied in psoriasis, were also identified. STAT1, EHF, FOSL1, STAT3, and NFKB1 were positively correlated with these DEGs in psoriasis lesions, whereas FOXC1 was negatively correlated with most DEGs. The decreased expression of the DEGs was accompanied by the downregulation of STAT1, EHF, FOSL1, STAT3, and NFKB1 and the upregulation of FOXC1 upon blocking interleukin 17 (IL-17) or tumor necrosis factor α signaling in psoriasis. Additionally, the downregulation of IL37 in psoriasis was negatively correlated with STAT1 and CXCL10, which are associated with Th1 responses. These results suggest that TFs play an important role in the pathogenesis of psoriasis, and interfering with the activity of key TFs may be a promising therapeutic strategy for psoriasis.
Collapse
Affiliation(s)
- Fanfan Zeng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbo Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China.,Department of Clinical Medicine, Lingui Clinical Medical College, Guilin Medical University, Guilin, China
| | - Di Lu
- Department of Clinical Medicine, Lingui Clinical Medical College, Guilin Medical University, Guilin, China
| | - Qianqian Liu
- Department of Clinical Medicine, Lingui Clinical Medical College, Guilin Medical University, Guilin, China
| | - Huoying Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China.,Department of Clinical Medicine, Lingui Clinical Medical College, Guilin Medical University, Guilin, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China.,NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|