1
|
Cheng C, Wang Y, Huo J, Zhang Y, Li R. SIRT6 knockdown alleviates keratinocyte hyperproliferation and inflammation in psoriasis via modulating acetylation of FOXO1. Int Immunopharmacol 2025; 146:113932. [PMID: 39733643 DOI: 10.1016/j.intimp.2024.113932] [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: 09/30/2024] [Revised: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 12/31/2024]
Abstract
The Sirtuins family (SIRT) has been implicated in numerous diseases, including psoriasis.However, the precise role of SIRT6 in psoriasis remains unclear. The analysis of publicly available RNA-seq data from GEO profiles showed that SIRT6 expression levels was significantly elevated in the lesional skins from patients with psoriasis, as compared to the non-lesional skins or the skins from normal healthy donors. It was also confirmed that SIRT6 and Ki67 expression was consistently upregulated inpsoriatic lesional skin,mouse models of psoriasis established by imiquimod treatment, and HaCat cells treated with M5. When SIRT6 was knocked down or inhibited in M5-treated HaCat cells, there was a significant suppression ofM5-induced increases in inflammatory cytokines such as interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. The upregulation of Ki67 expression and cell proliferation induced by M5 were also reduced. SIRT6 inhibitor also significantly reduced the epidermal thickness and Ki67 expression levels in mouse models of psoriasis. Mechanistically, SIRT6 knockdown or inhibition enhanced the nuclear translocation of forkhead box O 1 (FOXO1) by increasing its acetylation level. M5 treatment reduced the nuclear FOXO1 levels via enhancing the nuclear efflux of Foxo1. Knockdown or inhibition of SIRT6 resulted in an increase in nuclear FOXO1 levels, not through enhancing its nuclear influx, but possibly by impeding the nuclear efflux of Foxo1. In conclusion, the knockdown of the SIRT6 promoted the nuclear translocation of FOXO1 by upregulating its acetylation level, thereby inhibiting M5-induced hyperproliferation and inflammation of keratinocyte. Given the crucial role of SIRT6 in psoriasis, it may represent a promising target for the development of small-molecule inhibitors with therapeutic potential for psoriasis.
Collapse
Affiliation(s)
- Chuantao Cheng
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yuan Wang
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Jia Huo
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yanfei Zhang
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Ruilian Li
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
2
|
Sun Y, Wang J, Hu P, Tang Y, Wang Y, Ye J, Yang X, Yin J. Molecular mechanism through which Tripterygium hypoglaucum (Lévl.) Hutch alleviates psoriasis. Biomed Pharmacother 2024; 181:117647. [PMID: 39504627 DOI: 10.1016/j.biopha.2024.117647] [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: 09/10/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Tripterygium hypoglaucum (Lévl.) Hutch rhizome (THH) is mainly used in the clinical setting for the treatment of autoimmune diseases such as rheumatoid arthritis. In total, four active compounds were isolated from THH methanol extract (THH-MeOH)and identified. The HPLC results showed that the proportions of the active ingredients in THH-MeOH (i.e., celastrol, triptolide, and 3-O-acetyloleanolic acid) were 0.79 ‰, 0.46 ‰, and 0.76 ‰, respectively. THH-MeOH attenuated the M5-induced hyperproliferation of HaCaT cells, decreased the mRNA expression levels of inflammatory cytokines, and inhibited the phosphorylation of IκBα, NF-κB p65, MAPK, and STAT3/JAK2. Furthermore, THH-MeOH significantly reduced PASI scores in mice; reduced the level of Ki67 expression in skin tissues; decreased the expression of inflammatory cytokines in the skin lesions and serum; and ameliorated the IMQ-induced imbalance in the RORγt/Foxp3 ratio. The extract can attenuate psoriasis-like lesions by inhibiting cellular hyperproliferation, ameliorating inflammatory reactions, and modulating immune responses. Our work provides a theoretical basis to support the use of THH for treating psoriasis.
Collapse
Affiliation(s)
- Yumei Sun
- School of Ethnic Medicine, Yunnan Minzu University, Kunming, China; Yunnan Key Laboratory of Chiral Functional Substance Research and Application, Kunming, China; Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, China
| | - Jihong Wang
- School of Ethnic Medicine, Yunnan Minzu University, Kunming, China; Yunnan Key Laboratory of Chiral Functional Substance Research and Application, Kunming, China; Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, China
| | - Peiyao Hu
- School of Ethnic Medicine, Yunnan Minzu University, Kunming, China; Yunnan Key Laboratory of Chiral Functional Substance Research and Application, Kunming, China; Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, China
| | - Yi Tang
- School of Ethnic Medicine, Yunnan Minzu University, Kunming, China; Yunnan Key Laboratory of Chiral Functional Substance Research and Application, Kunming, China; Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, China
| | - Yanwen Wang
- School of Ethnic Medicine, Yunnan Minzu University, Kunming, China; Yunnan Key Laboratory of Chiral Functional Substance Research and Application, Kunming, China; Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, China
| | - Jianzhou Ye
- Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China.
| | - Xuesong Yang
- Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Junlin Yin
- School of Ethnic Medicine, Yunnan Minzu University, Kunming, China; Yunnan Key Laboratory of Chiral Functional Substance Research and Application, Kunming, China; Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, China.
| |
Collapse
|
3
|
Yanping H, Ting G, Xinzhu Z, Yaya L, Yuna Z, Qing L, Xueli M, Jing C. Yinxie I Formula attenuates imiquimod-induced psoriasis-like skin inflammation via IL-23/IL-17 axis. Arch Dermatol Res 2024; 316:540. [PMID: 39158742 PMCID: PMC11333517 DOI: 10.1007/s00403-024-03288-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/12/2024] [Accepted: 08/05/2024] [Indexed: 08/20/2024]
Abstract
Psoriasis is considered a chronic inflammatory skin disorder characterized by keratinocytes hyperproliferation. The IL-23/IL-17 immune pathway has been substantiated in numerous studies to be closely associated with psoriasis progression. Yinxie I Formula is a traditional Chinese medicine made from 9 herbal medicines, which has excellent clinical efficacy in psoriasis. However, to date, the mechanism of action of Yinxie I Formula against psoriasis remains unknown. In this perspective, we discuss the efficacy of Yinxie I Formula in mice with imiquimod (IMQ) induced psoriasis. Yinxie I Formula significantly reduced the area of skin lesions and the inflammatory response in mice with psoriasis. Furthermore, Yinxie I Formula alleviated the expression levels of inflammation-related genes IL-6, IL-17 A, IL-22, IL-23, TNF-α and IL-23, IL-18, IL-6 and IL-1β-related proteins and alleviated the abnormal surge of dendritic cells, macrophages and T cells in the skin and spleen. Meanwhile we found that Yinxie I Formula reduced the release of NO, TNF-α, IL-1β and IL-23 in lipopolysaccharide-induced mouse macrophage RAW264.7 cell line. The results suggest that the therapeutic mechanism of Yinxie I Formula may also be correlated with the STAT signaling pathway. We further analyzed the active ingredient of Yinxie I Formula, Buddleoside, which may be the main substance that exerts the therapeutic effect. In conclusion, we have investigated that Yinxie I Formula attenuates the IMQ-induced inflammatory response in psoriasis by inhibiting the IL-23/IL-17 axis, which lays the foundation for the antipsoriasis mechanism and provides a theoretical basis for the clinical promotion of Yinxie I Formula.
Collapse
Affiliation(s)
- He Yanping
- Department of Dermatology, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, PR China
| | - Gao Ting
- Department of Dermatology, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, PR China
| | - Zhou Xinzhu
- Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, PR China
| | - Lei Yaya
- Department of Dermatology, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, PR China
| | - Zhao Yuna
- Department of Dermatology, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, PR China
| | - Liu Qing
- Department of Dermatology, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, PR China
| | - Ma Xueli
- Department of Dermatology, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, PR China
| | - Chen Jing
- Department of Dermatology, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, PR China.
| |
Collapse
|
4
|
Yuan X, Ou C, Li X, Zhuang Z, Chen Y. The skin circadian clock gene F3 as a potential marker for psoriasis severity and its bidirectional relationship with IL-17 signaling in keratinocytes. Int Immunopharmacol 2024; 132:111993. [PMID: 38565044 DOI: 10.1016/j.intimp.2024.111993] [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: 01/23/2024] [Revised: 03/21/2024] [Accepted: 03/30/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE Psoriasis is an immune-mediated skin disease where the IL-17 signaling pathway plays a crucial role in its development. Chronic circadian rhythm disorder in psoriasis pathogenesis is gaining more attention. The relationship between IL and 17 signaling pathway and skin clock genes remains poorly understood. METHODS GSE121212 with psoriatic lesion and healthy controls was used as the exploration cohort for searching analysis. Datasets GSE54456, GSE13355, GSE14905, GSE117239, GSE51440, and GSE137218 were applied to validation analysis. Single-cell RNA sequencing (scRNA-seq) dataset GSE173706 was used to explore the F3 expression and related pathway activities in single-cell levels. Through intersecting with high-expression DEGs, F3 was selected as the signature skin circadian gene in psoriasis for further investigation. Functional analyses, including correlation analyses, prediction of transcription factors, protein-protein interaction, and single gene GSEA to explore the potential roles of F3. ssGSEA algorithm was performed to uncover the immune-related characteristics of psoriasis. We further explored F3 expression in the specific cell population in scRNA-seq dataset, besides this, AUCell analysis was performed to explore the pathway activities and the results were further compared between the specific cell cluster. Immunohistochemistry experiment, RT-qPCR was used to validate the location and expression of F3, small interfering RNA (siRNA) transfection experiment in HaCaT, and transcriptome sequencing analysis were applied to explore the potential function of F3. RESULTS F3 was significantly down-regulated in psoriasis and interacted with IL-17 signaling pathway. Low expression of F3 could upregulate the receptor of JAK-STAT signaling, thereby promoting keratinocyte inflammation. CONCLUSION Our research revealed a bidirectional link between the skin circadian gene F3 and the IL-17 signaling pathway in psoriasis, suggesting that F3 may interact with the IL-17 pathway by activating JAK-STAT within keratinocytes and inducing abnormal intracellular inflammation.
Collapse
Affiliation(s)
- Xiuqing Yuan
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Caixin Ou
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Xinhui Li
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Zhe Zhuang
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Yongfeng Chen
- Dermatology Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Liu W, Wang Y, Zhang Y, Zhou M, Gu H, Lu M, Xia Y. Rh family C glycoprotein contributes to psoriatic inflammation through regulating the dysdifferentiation and cytokine secretion of keratinocytes. J Dermatol Sci 2024; 114:2-12. [PMID: 38514279 DOI: 10.1016/j.jdermsci.2024.02.007] [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: 08/24/2023] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Keratinocyte dysdifferentiation and proinflammatory cytokine production play a central role in psoriatic inflammation. According to recent studies, the Rh family C glycoprotein (RHCG) enhances cell proliferation and disrupts cell differentiation. However, the specific role of RHCG psoriasis development remains unclear. OBJECTIVE We here explored the effect of RHCG on keratinocytes under psoriatic inflammation. METHODS The cell counting kit‑8 assay was conducted to assess proliferation. RHCG protein expression was assessed through western blotting and enzyme-linked immunosorbent assays. The expression of proinflammatory cytokines and differentiation markers was analyzed through a quantitative reverse-transcription polymerase chain reaction. RESULTS Both RHCG mRNA and protein levels increased in psoriatic skin. Notably, cultured keratinocytes treated with an M5 cocktail, which mimics psoriatic inflammation, exhibited higher RHCG expression. Furthermore, RHCG overexpression promoted keratinocyte proliferation, accompanied by an increase in the production of interleukin (IL)-1β, IL-6, and IL-8, and tumor necrosis factor-α. RHCG overexpression also resulted in higher expression of keratin 17, a differentiation marker. Conversely, RHCG gene knockdown reduced keratinocyte proliferation and cytokine secretion. RHCG inhibition in cells recovered both keratin 1 and loricrin expression. Additionally, RHCG overexpression facilitated the phosphorylation of nuclear factor-kappa B and extracellular signal-regulated protein kinase signaling pathways. Importantly, when these signaling pathways were inhibited, the effect of RHCG on keratinocytes was attenuated. CONCLUSION These findings support the substantial role of RHCG in psoriatic inflammation development and suggest that RHCG serves as a potential target for psoriasis treatment.
Collapse
Affiliation(s)
- Wei Liu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yaqi Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yitian Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mingzhu Zhou
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hanjiang Gu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mei Lu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
6
|
Chen S, Yu R, Zhao F, Sun L, Yin Y, Zhang G, Chen Q, Shu Q. Network pharmacology and molecular docking to explore the mechanism of a clinical proved recipe for external use of clearing heat and removing dampness in the treatment of immune-related cutaneous adverse events. Medicine (Baltimore) 2024; 103:e37504. [PMID: 38489696 PMCID: PMC10939542 DOI: 10.1097/md.0000000000037504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/04/2023] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Immune-related cutaneous adverse events (ircAEs) will undermine the patients' quality of lives, and interrupt the antitumor therapy. A clinical proved recipe for external use of clearing heat and removing dampness (Qing-Re-Li-Shi Formula, hereinafter referred to as "QRLSF") is beneficial to the treatment of ircAEs in clinical practice. Our study will elucidate the mechanism of QRLSF against ircAEs based on network pharmacology and molecular docking. The active components and corresponding targets of QRLSF were collected through traditional Chinese medicine systems pharmacology database. GeneCards, online Mendelian inheritance in man, and pharmacogenomics knowledgebase were used to screen the targets of ircAEs. The intersecting targets between drug and disease were acquired by venn analysis. Cytoscape software was employed to construct "components-targets" network. Search tool for the retrieval of interacting genes/proteins database was applied to establish the protein-protein interaction network and then its core targets were identified. Gene ontology and Kyoto encyclopedia of genes and genomes analysis was performed to predict the mechanism. The molecular docking verification of key targets and related phytomolecules was accomplished by AutoDock Vina software. Thirty-nine intersecting targets related to QRLSF against ircAEs were recognized. The analysis of network clarified 5 core targets (STAT3, RELA, TNF, TP53, and NFKBIA) and 4 key components (quercetin, apigenin, luteolin, and ursolic acid). The activity of QRLSF against ircAEs could be attributed to the regulation of multiple biological effects via multi-pathways (PI3K-Akt pathway, cytokine-cytokine receptor interaction, JAK-STAT pathway, chemokine pathway, Th17 cell differentiation, IL-17 pathway, TNF pathway, and Toll-like receptor pathway). The binding activities were estimated as good level by molecular docking. These discoveries disclosed the multi-component, multi-target, and multi-pathway characteristics of QRLSF against ircAEs, providing a new strategy for such medical problem.
Collapse
Affiliation(s)
- Shuyi Chen
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Rui Yu
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Fangmin Zhao
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lin Sun
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yudan Yin
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Gaochenxi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Qunwei Chen
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Qijin Shu
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Pan J, Wu J, Zhang S, Wang K, Ji G, Zhou W, Dang Y. Targeted metabolomics revealed the mechanisms underlying the role of Liansu capsule in ameliorating functional dyspepsia. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117568. [PMID: 38092317 DOI: 10.1016/j.jep.2023.117568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/26/2023] [Accepted: 12/06/2023] [Indexed: 12/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liansu capsule could alleviate dyspeptic symptoms; however, the mechanisms underlying its role in treating functional dyspepsia (FD) remain unclear. AIM OF THE STUDY To elucidate the mechanism underlying the efficacy of Liansu capsule in alleviating FD symptoms. MATERIALS AND METHODS Thirty-six male mice were randomly divided into the following six groups: control, model, low-strength Liansu, moderate-strength Liansu, high-strength Liansu, and domperidone groups. Small intestine propulsion rate, gastric residual rate and histopathological analysis were performed to evaluate efficacy of Liansu capsule. Levels of interleukin-1β, interleukin-6, tumor necrosis factor α, phosphorylation of p65, ghrelin and gastrin were verified by real-time quantitative polymerase chain reaction and immunofluorescence assays. Targeted metabolomic analyses, western blotting and immunofluorescence assays were used to explore the mechanism of Liansu capsule in ameliorating FD. RESULTS The Liansu capsule significantly ameliorated the symptoms of FD, and markedly increased the levels of ghrelin and gastrin. Moreover, Liansu capsule significantly downregulated the levels of the proinflammatory cytokine interleukin-1β, interleukin-6, tumor necrosis factor α, and inhibited the phosphorylation of p65. Targeted metabolomic analyses showed that Liansu capsule significantly reduced the levels of deoxycholic acid and hyodeoxycholic acid, which were significantly elevated in the model group. Furthermore, these results showed that deoxycholic acid and hyodeoxycholic acid markedly promoted the levels of Takeda G-protein-coupled receptor 5 (TGR5), phosphorylated signal transducer and activator of transcription 3 (STAT3), and Kruppel-like factor 5 (KLF5) in vitro. whereas, Liansu capsule significantly reduced the levels of TGR5, phosphorylated STAT3, and KLF5. CONCLUSION Our findings indicated that Liansu capsule improved FD by regulating the deoxycholic acid/hyodeoxycholic acid-TGR5-STAT3-KLF5 axis. The findings reveal a novel mechanism underlying the role of Liansu capsule, which may be a promising therapeutic strategy for FD.
Collapse
Affiliation(s)
- Jiashu Pan
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Jiaxuan Wu
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Shengan Zhang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Kai Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Yanqi Dang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
8
|
Liu S, Liu F, Zhang Z, Zhuang Z, Chen Y. PTPN2 inhibits the proliferation of psoriatic keratinocytes by dephosphorylation of STAT3. Cell Biochem Funct 2024; 42:e3947. [PMID: 38379221 DOI: 10.1002/cbf.3947] [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: 08/21/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/22/2024]
Abstract
Psoriasis is a recurrent and protracted disease that severely impacts the patient's physical and mental health. Thus, there is an urgent need to explore its pathogenesis to identify therapeutic targets. The expression level of protein tyrosine phosphatase nonreceptor type 2 (PTPN2) was analyzed by immunohistochemistry techniques in psoriatic tissues and imiquimod-induced psoriatic mouse models. PTPN2 and signal transducer and activator of transcription 3 (STAT3) were overexpressed or silenced in human keratinocytes or an interleukin (IL)-6-induced psoriasis HaCaT cell model using overexpression plasmid transfection or small interfering RNA technology in vitro, and the effects of PTPN2 on STAT3, HaCaT cell function, and autophagy levels were investigated using reverse transcription-quantitative polymerase chain reaction, Western blot, Cell Counting Kit 8, 5-ethynyl-20-deoxyuridine, flow cytometry, and transmission electron microscopy. PTPN2 expression was found to be significantly downregulated in psoriatic tissues. Then, the in vitro antipsoriatic properties of PTPN2 were investigated in an IL-6-induced psoriasis-like cell model, and the results demonstrated that inhibition of keratinocyte proliferation by PTPN2 may be associated with elevated STAT3 dephosphorylation and autophagy levels. These findings provide novel insights into the mechanisms of autophagy in psoriatic keratinocytes and may be essential for developing new therapeutic strategies to improve inflammatory homeostasis in psoriatic patients.
Collapse
Affiliation(s)
- Shougang Liu
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Fanghua Liu
- Department of Dermatology, Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
- Department of Dermatology, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, People's Republic of China
| | - Zeqiao Zhang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Zhe Zhuang
- Department of Dermatology, Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| | - Yongfeng Chen
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Department of Dermatology, Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| |
Collapse
|
9
|
Srikanth M, Rasool M. Resistin - A Plausible Therapeutic Target in the Pathogenesis of Psoriasis. Immunol Invest 2024; 53:115-159. [PMID: 38054436 DOI: 10.1080/08820139.2023.2288836] [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: 12/07/2023]
Abstract
Resistin, a cytokine hormone predominantly secreted by adipose tissue, is elevated in various metabolic disorders such as obesity, type 2 diabetes, and cardiovascular disease. In addition to its involvement in metabolic regulation, resistin has been implicated in the pathogenesis of psoriasis, a chronic inflammatory skin disorder. Numerous studies have reported increased resistin levels in psoriatic skin lesions, suggesting a possible association between resistin and psoriasis. Recent studies have suggested the potential involvement of resistin in the development and progression of certain cancers. Resistin is overexpressed in breast, colorectal, and gastric cancers. This suggests that it may play a role in the development of these cancers, possibly by inducing inflammation and cell growth. The link between resistin and cancer raises the possibility of shared underlying mechanisms driving the pathogenesis of psoriasis. Chronic inflammation, one such mechanism, is a hallmark of psoriasis and cancer. Further research is needed to fully understand the relationship between resistin and psoriasis. Identifying potential therapeutic targets is crucial for effective management of psoriasis. By doing so, we may be able to develop more effective treatment options for individuals living with psoriasis and ultimately improve their quality of life. Ultimately, a more comprehensive understanding of the mechanisms underlying the impact of resistin on psoriasis is essential for advancing our knowledge and finding new ways to treat and manage this challenging condition.
Collapse
Affiliation(s)
- Manupati Srikanth
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - Mahaboobkhan Rasool
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| |
Collapse
|
10
|
Chen Y, Xiang Y, Miao X, Kuai L, Ding X, Ma T, Li B, Fan B. METTL14 promotes IL-6-induced viability, glycolysis and inflammation in HaCaT cells via the m6A modification of TRIM27. J Cell Mol Med 2024; 28:e18085. [PMID: 38146129 PMCID: PMC10844716 DOI: 10.1111/jcmm.18085] [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: 06/29/2023] [Revised: 11/29/2023] [Accepted: 12/10/2023] [Indexed: 12/27/2023] Open
Abstract
Interleukin-6 (IL-6) is a cytokine generated by healthy constituents of the skin, but is also up-regulated by a wide range of skin lesions and inflammatory conditions to trigger cytopathy of skin cells. TRIM27 was identified to contribute to the functional effects of IL-6 on skin cells. However, the underlying mechanism was not clear. Lentivirus infection was used for gene overexpression or silencing. RT-PCR and Western blot were used to respectively assess mRNA and protein levels. Cell viability was assessed by CCK-8 assay. Extracellular flux analysis was used to assess the levels of oxygen consumption rate and extracellular acidification rate. Mouse back skin was treated with imiquimod to produce psoriasis-like inflammation in vivo. Histological assessment and immunohistochemistry staining were respectively applied to analyse lesioned mouse and human skin samples. IL-6-induced increased viability, glycolysis and inflammation in keratinocytes was inhibited both by a chemical methylation inhibitor and by METTL14 knockdown. Further investigation found that METTL14 induces m6A methylation of TRIM27, which is recognized by a m6A reader, IGF2BP2. Elevation of TRIM27 level and activation of IL-6/STAT3 signalling pathway were found in an in vivo psoriasis-like inflammation model, whereas inhibition m6A methylation strongly alleviated the inflammation. Finally, METTL14, TRIM27, STAT3, p-STAT3 and IL-6 expressions were all found to be increased in clinical skin samples of psoriatic patients. Our results unravelled METTL14/TRIM27/IGF2BP2 signalling axis in keratinocyte cytopathy, which plays a critical role in facilitating the activation of IL-6/STAT3 signalling pathway. Our findings should provide inspirations for the design of new therapeutics for skin inflammatory diseases including psoriasis.
Collapse
Affiliation(s)
- Yiran Chen
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yanwei Xiang
- Engineering Research Center of Traditional Chinese Medicine Intelligent RehabilitationMinistry of EducationShanghaiChina
- School of Rehabilitation ScienceShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xiao Miao
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Innovation Research Institute of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xiaojie Ding
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Tian Ma
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Bin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Bin Fan
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
11
|
Meng Q, Liu Y, Yao L, Ma Z, Guo L, Hu T, Luo Y, Chen J, Dang E, Li Z. Serine deficiency exacerbates psoriatic skin inflammation by regulating S-adenosyl methionine-dependent DNA methylation and NF-κB signalling activation in keratinocytes. J Eur Acad Dermatol Venereol 2024; 38:145-156. [PMID: 37669859 DOI: 10.1111/jdv.19492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/03/2023] [Indexed: 09/07/2023]
Abstract
BACKGROUND Serine metabolism is crucial for tumour oncogenesis and immune responses. S-adenosyl methionine (SAM), a methyl donor, is typically derived from serine-driven one-carbon metabolism. However, the involvement of serine metabolism in psoriatic skin inflammation remains unclear. OBJECTIVES To investigate the association between serine metabolism and psoriatic skin inflammation. METHODS Clinical samples were collected from patients with psoriasis and the expression of serine biosynthesis enzymes was evaluated. The HaCaT human keratinocyte cell line was transfected with small interfering RNA (siRNA) of key enzyme or treated with inhibitors. RNA sequencing and DNA methylation assays were performed to elucidate the mechanisms underlying serine metabolism-regulated psoriatic keratinocyte inflammation. An imiquimod (IMQ)-induced psoriasis mouse model was established to determine the effect of the SAM administration on psoriatic skin inflammation. RESULTS The expression of serine synthesis pathway enzymes, including the first rate-limiting enzyme in serine biosynthesis, phosphoglycerate dehydrogenase (PHGDH), was downregulated in the epidermal lesions of patients with psoriasis compared with that in healthy controls. Suppressing PHGDH in keratinocytes promoted the production of proinflammatory cytokines and enrichment of psoriatic-related signalling pathways, including the tumour necrosis factor-alpha (TNF-α) signalling pathway, interleukin (IL)-17 signalling pathway and NF-κB signalling pathway. In particular, PHGDH inhibition markedly promoted the secretion of IL-6 in keratinocytes with or without IL-17A, IL-22, IL-1α, oncostatin M and TNF-α (mix) stimulation. Mechanistically, PHGDH inhibition upregulated the expression of IL-6 by inhibiting SAM-dependent DNA methylation at the promoter and increasing the binding of myocyte enhancer factor 2A. Furthermore, PHGDH inhibition increased the secretion of IL-6 by increasing the activation of NF-κB via SAM inhibition. SAM treatment effectively alleviated IMQ-induced psoriasis-like skin inflammation in mice. CONCLUSIONS Our study revealed the crucial role of PHGDH in antagonising psoriatic skin inflammation and indicated that targeting serine metabolism may represent a novel therapeutic strategy for treating psoriasis.
Collapse
Affiliation(s)
- Qinqin Meng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Liu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Leiqing Yao
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhimiao Ma
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lu Guo
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ting Hu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yixin Luo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiaoling Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhengxiao Li
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
12
|
Zheng JQ, Zhan Y, Huang WJ, Chen ZY, Wu WH. N6-methyladenosine of TRIM27 enhances the stem cell-type phenotype of cisplatin-resistant colorectal cancer cells. Biochem Biophys Rep 2023; 36:101572. [PMID: 38024865 PMCID: PMC10658205 DOI: 10.1016/j.bbrep.2023.101572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Colorectal cancer (CRC), classified as a lethal form of cancer, substantially threatens human well-being. Cancer stem cells (CSCs) reflect subsets for cancerous cells having basic stem-cell type properties, being significantly involved in the development of chemoresistance and tumor relapsing. The aberrant TRIM27 expression in various types of cancer indicates its potential involvement in cancer growth and progression. The current understanding of the TRIM27 involvement in CRC remains limited. In current study indicated that TRIM27 can potentially promote CSC-type phenotype of Cisplatin (DDP)-resistant CRC cells. YTHDF1 recruitment onto m6A-amended TRIM27 was crucial for facilitating the TRIM27 translating process in DDP-resistant CRC cells. The present research proposes that TRIM27 exhibits an oncogenic role by enhancing the CSC-type properties in DDP-resistant CRC via the m6A-modified pathway. The potential therapy for combating the relapse of CRC may include TRIM27 and YTHDF1, as they have been found to have significant roles in promoting CSC-type phenotypic characteristics.
Collapse
Affiliation(s)
- Jun-qiong Zheng
- Department of Medical Oncology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, Fujian, China
| | - Ying Zhan
- Department of Medical Oncology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, Fujian, China
| | - Wen-jing Huang
- Department of Medical Oncology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, Fujian, China
| | - Zhi-yong Chen
- Department of Medical Oncology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, Fujian, China
| | - Wei-hao Wu
- Department of Medical Oncology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, Fujian, China
| |
Collapse
|
13
|
Zhuang X, Liu T, Wei L, Gao Y, Gao J. RNA sequencing reveals the mechanism of FTO in inhibiting inflammation and excessive proliferation of lipopolysaccharide-induced human glomerular mesangial cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3835-3846. [PMID: 37358794 DOI: 10.1007/s00210-023-02570-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/06/2023] [Indexed: 06/27/2023]
Abstract
Chronic glomerulonephritis (CGN) is a leading cause of end-stage renal disease in China; thus, there is an urgent need for effective therapeutic targets and strategies for CGN treatment. However, studies on CGN pathogenesis are limited. In this study, we found that the fat mass and obesity-associated protein (FTO) was significantly decreased in the lipopolysaccharide (LPS)-induced human glomerular mesangial cells (HGMCs) (P < 0.01) and kidney tissues of CGN patients (P < 0.05). Moreover, double-labeling immunofluorescence and flow cytometry assays demonstrated that the overexpression of FTO could inhibit inflammation and excessive proliferation of HGMCs. Furthermore, RNA-sequencing (RNA-seq) and real-time quantitative polymerase chain reaction (RT-qPCR) analyses revealed that FTO overexpression induced differential expression of 269 genes (absolute fold change ≥ 2 and P-value < 0.05), including 143 upregulated and 126 downregulated genes. Further functional analysis of these differentially expressed genes by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses suggested that FTO possibly mediates its inhibitory function by regulating the mammalian target of rapamycin (mTOR) signaling pathway and substance metabolism. Lastly, analysis of the PPI network and further identification of the top 10 hub genes (RPS15, RPS18, RPL18A, GNB2L1, RPL19, EEF1A1, RPS25, FAU, UBA52, and RPS6) indicated that FTO mediates its function by affecting the ribosomal proteins. Therefore, in this study, we elucidated the important role of FTO in the regulation of inflammation and excessive proliferation of HGMCs, suggesting FTO administration as a suitable therapeutic intervention for CGN.
Collapse
Affiliation(s)
- Xingxing Zhuang
- Department of Pharmacy, Chaohu Hospital of Anhui Medical University, No. 64 North Chaohu Road, Chaohu, 238000, Anhui, China
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Hefei, 230012, Anhui, China
| | - Tao Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Hefei, 230012, Anhui, China
| | - Liangbing Wei
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Hefei, 230012, Anhui, China
| | - Yachen Gao
- Department of Nephropathy, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Hefei, 230012, Anhui, China
| | - Jiarong Gao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Hefei, 230012, Anhui, China.
| |
Collapse
|
14
|
Tu Z, Wei W, Xiang Q, Wang W, Zhang S, Zhou H. Pro-inflammatory cytokine IL-6 regulates LMO4 expression in psoriatic keratinocytes via AKT/STAT3 pathway. Immun Inflamm Dis 2023; 11:e1104. [PMID: 38156380 PMCID: PMC10698831 DOI: 10.1002/iid3.1104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/03/2023] [Accepted: 11/18/2023] [Indexed: 12/30/2023] Open
Abstract
The transcription factor LIM-only protein 4 (LMO4) is overexpressed in the psoriatic epidermis and regulates keratinocyte proliferation and differentiation. High LMO4 expression levels are induced by interleukin-23 (IL-23) to activate the AKT/STAT3 signaling pathway. Interleukin-6 (IL-6) is mainly involved in regulating T cell functions and development in patients with psoriasis. However, whether LMO4 expression is regulated by IL-6 remains unclear. Therefore, the purpose of this study is to explore the role and molecular mechanisms of IL-6 in regulating LMO4 expression. The interleukin-6 (IL-6) levels in human plasma were determined using a chemiluminescence immunoassay system. A psoriasis-like mouse model was established using imiquimod induction. Epidermal keratinocytes (HaCaT) were cultured in defined keratinocyte-serum-free medium and stimulated by IL-6 alone or with inhibitors. The proteins of interest were detected using western blot analysis, immunofluorescence, and immunohistochemistry. The 5-ethynyl-2'-deoxyuridine assay was used to detect cell proliferation. The results revealed that IL-6 levels were markedly increased in the plasma of patients with psoriasis, compared to healthy control. The high expression of LMO4 was consistent with high levels of IL-6, p-AKT, and p-STAT3 in the lesions of both psoriasis patients and imiquimod-induced psoriasis-like mice. IL-6 activates the AKT/STAT3 signaling pathway, followed by LMO4 high-expression in HaCaT cells. IL-6 induces HaCaT proliferation and differentiation via AKT/STAT3 signaling pathway activation. We think that the high expression of LMO4 in psoriatic keratinocytes requires IL-6 to activate the AKT/STAT3 signaling pathway and leads to epidermal keratinocytes abnormal proliferation and differentiation.
Collapse
Affiliation(s)
- Zhenzhen Tu
- Department of Immunology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Wei Wei
- Department of DermatologyAffiliated Provincial Hospital of Anhui Medical UniversityHefeiChina
| | - Qiantong Xiang
- Department of DermatologySecond People's Hospital of Hefei Affiliated of Anhui Medical UniversityHefeiChina
| | - Wenwen Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
| | - Siping Zhang
- Department of DermatologyAffiliated Provincial Hospital of Anhui Medical UniversityHefeiChina
| | - Haisheng Zhou
- Department of Immunology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
- Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesAnhui Medical UniversityHefeiChina
- The Center for Scientific Research of Anhui Medical UniversityHefeiChina
- The Institute of DermatologyAnhui Medical UniversityHefeiChina
| |
Collapse
|
15
|
He J, Peng F, Chang J, Zhao Y, Qu Y, Liu J, Liu R, Li P, Cai G, Hong Q, Chen X. The therapeutic effect of Shenhua tablet against mesangial cell proliferation and renal inflammation in mesangial proliferative glomerulonephritis. Biomed Pharmacother 2023; 165:115233. [PMID: 37536037 DOI: 10.1016/j.biopha.2023.115233] [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: 04/23/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/05/2023] Open
Abstract
Shenhua tablet (SH), a formulation of traditional Chinese medicine, exerts renoprotective effect on chronic kidney diseases, and it has been found to restrain inflammation, but the mechanism is still unclear. Here, we explored the potential renoprotection of SH in mesangial proliferative glomerulonephritis (MsPGN) rat model induced by anti-Thy1 antibody. Administration of SH reduced urinary albumin/creatinine ratio (UACR) and significantly attenuated mesangial cell proliferation and renal inflammation. Notably, SH protected rats against renal inflammation, which was associated with decreasing macrophage infiltration and promoting macrophage anti-inflammatory activity. Network analysis combined with arrays identified the Janus kinase signal transducer and activator of transcription (JAK-STAT) signaling pathway as the main pathways of SH could target inflammation. Furthermore, it was confirmed that mesangial cell proliferation, which response to inflammation, were alleviated by ASS1 expression enhanced after SH administration both in vivo and in vitro. Collectively, SH has the beneficial on relieving the progression of MsPGN to alleviate inflammation and mesangial proliferation by inhibiting STAT3 phosphorylation and maintains the expression level of ASS1, might be an effective strategy for treating MsPGN.
Collapse
Affiliation(s)
- Jiayi He
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Fei Peng
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Jiakai Chang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yinghua Zhao
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, China
| | - Yilun Qu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Jiaona Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Ran Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Quan Hong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China; School of Medicine, Nankai University, Tianjin 300071, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
16
|
Cai Z, Zeng Y, Liu Z, Zhu R, Wang W. Curcumin Alleviates Epidermal Psoriasis-Like Dermatitis and IL-6/STAT3 Pathway of Mice. Clin Cosmet Investig Dermatol 2023; 16:2399-2408. [PMID: 37675183 PMCID: PMC10478781 DOI: 10.2147/ccid.s423922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/25/2023] [Indexed: 09/08/2023]
Abstract
Background To further investigate why curcumin (CUR) can attenuate psoriasis-like dermatitis of mice. Methods and Results Sixteen mice were randomized into four groups. The control group used carrier cream, and the model and the CUR group were applied with topical 5% imiquimod in the naked mice skin once a day for 6 days (62.5 mg/day/mice). Meanwhile, the control and model mice were given the same dose of saline by oral means, while mice in the CUR groups received oral drug doses of 50 and 100 mg/kg once a day for 6 days, respectively. CUR could largely improve imiquimod-induced lesions of mice. By using the ELISA and qPCR, we found that the protein and mRNA levels of epidermal TNF-α and IL-6 were inhibited by CUR. The phosphorylation levels of STAT3 and its downstream associated protein levels (eg, Cyclin D1, Bcl-2 and Pim1) in skin tissues of different groups were also inhibited by CUR. Furthermore, the results of immunohistochemistry also showed the repressed effect of CUR for the expression of TNF-α, IL-6 and p-STAT3 in psoriasis-like lesions of mice. Conclusion CUR can effectively ameliorate the featured lesions of psoriasis mice, which may be closely associated with the involvement of IL-6/STAT3 signaling.
Collapse
Affiliation(s)
- Zhenguo Cai
- Department of Dermatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Yibin Zeng
- Department of Dermatology, Minhang Hospital, Fudan University/Central Hospital of Minhang District, Shanghai, 201199, People’s Republic of China
| | - Zhuohang Liu
- Department of Dermatology, Minhang Hospital, Fudan University/Central Hospital of Minhang District, Shanghai, 201199, People’s Republic of China
| | - Ruizheng Zhu
- Department of Dermatology, Minhang Hospital, Fudan University/Central Hospital of Minhang District, Shanghai, 201199, People’s Republic of China
| | - Wuqing Wang
- Department of Dermatology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
- Department of Dermatology, Minhang Hospital, Fudan University/Central Hospital of Minhang District, Shanghai, 201199, People’s Republic of China
| |
Collapse
|
17
|
Słuczanowska-Głabowska S, Salmanowicz M, Staniszewska M, Pawlik A. The Role of Sirtuins in the Pathogenesis of Psoriasis. Int J Mol Sci 2023; 24:10782. [PMID: 37445960 DOI: 10.3390/ijms241310782] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 06/25/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Psoriasis is the most common chronic inflammatory skin disease with a genetic basis. It is characterised by keratinocyte hyperproliferation, parakeratosis and inflammatory cell infiltration. Psoriasis negatively affects a patient's physical and emotional quality of life. Sirtuins (SIRTs; silent information regulators) are an evolutionarily conserved group of enzymes involved in the post-translational modification of proteins, including deacetylation, polyADP-ribosylation, demalonylation and lipoamidation. SIRTs are involved in a number of cellular pathways related to ageing, inflammation, oxidative stress, epigenetics, tumorigenesis, the cell cycle, DNA repair and cell proliferation, positioning them as an essential component in the pathogenesis of many diseases, including psoriasis. Activation of SIRT1 counteracts oxidative-stress-induced damage by inhibiting the mitogen-activated protein kinases (MAPK), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and signal transducer and activator of transcription 3 (STAT3) pathways and may mitigate pathological events in psoriasis. There is a significant reduction in the expression of SIRT1, SIRT2, SIRT3, SIRT4 and SIRT5 and an increase in the expression of SIRT6 and SIRT7 in psoriasis. The aim of the review is to draw the attention of physicians and scientists to the importance of SIRTs in dermatology and to provide a basis and impetus for future discussions, research and pharmacological discoveries to modulate SIRT activity. In light of the analysis of the mode of action of SIRTs in psoriasis, SIRT1-SIRT5 agonists and SIRT6 and SIRT7 inhibitors may represent new therapeutic options for the treatment of psoriasis.
Collapse
Affiliation(s)
| | - Maria Salmanowicz
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland
| | - Marzena Staniszewska
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland
| |
Collapse
|
18
|
Tan Y, Zhang F, Fan X, Lu S, Liu Y, Wu Z, Huang Z, Wu C, Cheng G, Li B, Huang J, Stalin A, Zhou W, Wu J. Exploring the effect of Yinzhihuang granules on alcoholic liver disease based on pharmacodynamics, network pharmacology and molecular docking. Chin Med 2023; 18:52. [PMID: 37165407 PMCID: PMC10173499 DOI: 10.1186/s13020-023-00759-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/24/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Yinzhihuang granules (YZHG) is a commonly used Chinese patent medicine for the treatment of liver disease. However, the mechanism of YZHG in alcoholic liver disease (ALD) is still unclear. METHODS This study combined liquid chromatography-mass spectrometry technology, pharmacodynamics, network pharmacology and molecular docking methods to evaluate the potential mechanism of YZHG in the treatment of ALD. RESULTS A total of 25 compounds including 4-hydroxyacetophenone, scoparone, geniposide, quercetin, baicalin, baicalein, chlorogenic acid and caffeic acid in YZHG were identified by ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). The pharmacodynamic investigations indicated that YZHG could improve liver function and the degree of liver tissue lesions, and reduce liver inflammation and oxidative stress in ALD mice. Network pharmacology analysis showed that YZHG treated ALD mainly by regulating inflammation-related signaling pathways such as the PI3K-Akt signaling pathway. The results of the PPI network and molecular docking showed that the targets of SRC, HSP90AA1, STAT3, EGFR and AKT1 could be the key targets of YZHG in the treatment of ALD. CONCLUSION This study explored the potential compounds, potential targets and signaling pathways of YZHG in the treatment of ALD, which is helpful to clarify the efficacy and mechanism of YZHG and provide new insights for the clinical application of YZHG.
Collapse
Affiliation(s)
- Yingying Tan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fanqin Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotian Fan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shan Lu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhishan Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhihong Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guoliang Cheng
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Bing Li
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi, China
| | - Jiaqi Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China.
| | - Wei Zhou
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
19
|
Han D, Gong H, Wei Y, Xu Y, Zhou X, Wang Z, Feng F. Hesperidin inhibits lung fibroblast senescence via IL-6/STAT3 signaling pathway to suppress pulmonary fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154680. [PMID: 36736168 DOI: 10.1016/j.phymed.2023.154680] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/19/2022] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and fatal lung disease with obscure pathogenesis. Increasing evidence suggests that cellular senescence is an important mechanism underlying in IPF. Clinical treatment with drugs, such as pirfenidone and nintedanib, reduces the risk of acute exacerbation and delays the decline of pulmonary function in patients with mild to moderate pulmonary fibrosis, and with adverse reactions. Hesperidin was previously shown to alleviate pulmonary fibrosis in rats by attenuating the inflammation response. Our previous research indicated that the Citrus alkaline extracts, hesperidin as the main active ingredient, could exert anti-pulmonary fibrosis effects by inhibiting the senescence of lung fibroblasts. However, whether hesperidin could ameliorate pulmonary fibrosis by inhibiting fibroblast senescence needed further study. PURPOSE This work aimed to investigate whether and how hesperidin can inhibit lung fibroblast senescence and thereby alleviate pulmonary fibrosis METHODS: Bleomycin was used to establish a mouse model of pulmonary fibrosis and doxorubicin was used to establish a model of cellular senescence in MRC-5 cells in vitro. The therapeutic effects of hesperidin on pulmonary fibrosis using haematoxylin-eosin staining, Masson staining, enzyme-linked immunosorbent assay, immunohistochemistry, western blotting and quantitative Real-Time PCR. The anti-senescent effect of hesperidin in vivo and in vitro was assessed by western blotting, quantitative Real-Time PCR and senescence-associated β-galactosidase RESULTS: We demonstrated that hesperidin could alleviate bleomycin-induced pulmonary fibrosis in mice. The expression level of senescence marker proteins p53, p21, and p16 was were downregulated, along with the myofibroblast marker α-SMA. The number of senescence-associated β-galactosidase-positive cells was significantly reduced by hesperidin intervention in vivo and in vitro. In addition, hesperidin could inhibit the IL6/STAT3 signaling pathway. Furthermore, suppression of the IL-6/STAT3 signaling pathway by pretreatment with the IL-6 inhibitor LMT-28 attenuating effect of hesperidin on fibroblast senescence in vitro. CONCLUSIONS These data illustrated that hesperidin may be potentially used in the treatment of IPF based on its ability to inhibit lung fibroblast senescence.
Collapse
Affiliation(s)
- Di Han
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Haiying Gong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China
| | - Yun Wei
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yong Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China; School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xianmei Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| | - Zhichao Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| | - Fanchao Feng
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| |
Collapse
|
20
|
Li N, Lin G, Zhang H, Sun J, Gui M, Liu Y, Li W, Zhan Z, Li Y, Pan S, Liu J, Tang J. Lyn attenuates sepsis-associated acute kidney injury by inhibition of phospho-STAT3 and apoptosis. Biochem Pharmacol 2023; 211:115523. [PMID: 37003346 DOI: 10.1016/j.bcp.2023.115523] [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: 12/20/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023]
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is a life-threatening condition associated with high mortality and morbidity. However, the underlying pathogenesis of SA-AKI is still unclear. Lyn belongs to Src family kinases (SFKs), which exert numerous biological functions including modulation in receptor-mediated intracellular signaling and intercellular communication. Previous studies demonstrated that Lyn gene deletion obviously aggravates LPS-induced lung inflammation, but the role and possible mechanism of Lyn in SA-AKI have not been reported yet. Here, we found that Lyn protected against renal tubular injury in cecal ligation and puncture (CLP) induced AKI mouse model by inhibition of signal transducer and activator of transcription 3 (STAT3) phosphorylation and cell apoptosis. Moreover, Lyn agonist MLR-1023 pretreatment improved renal function, inhibited STAT3 phosphorylation and decreased cell apoptosis. Thus, Lyn appears to play a crucial role in orchestrating STAT3-mediated inflammation and cell apoptosis in SA-AKI. Hence, Lyn kinase may be a promising therapeutic target for SA-AKI.
Collapse
Affiliation(s)
- Nannan Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Guoxin Lin
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Jian Sun
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Ming Gui
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Yan Liu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Wei Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Zishun Zhan
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Yisu Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Shiqi Pan
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Jishi Liu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Juan Tang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
21
|
Wang C, Zhang P, Li Y, Wang X, Guo L, Li J, Jiao H. Downregulation of TRIM27 alleviates hypoxic-ischemic encephalopathy through inhibiting inflammation and microglia cell activation by regulating STAT3/HMGB1 axis. J Chem Neuroanat 2023; 129:102251. [PMID: 36796734 DOI: 10.1016/j.jchemneu.2023.102251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
TRIM27 expression was increased in the Parkinson's disease (PD), and knockdown of TRIM27 in PC12 cells significantly inhibited cell apoptosis, indicating that downregulation of TRIM27 exerts a neuroprotective effect. Herein, we investigated TRIM27 role in hypoxic-ischemic encephalopathy (HIE) and the underlying mechanisms. HIE models were constructed in newborn rats using hypoxic ischemic (HI) treatment and PC-12/BV2 cells with oxygen glucose deprivation (OGD), respectively. The results demonstrated that TRIM27 expression was increased in the brain tissues of HIE rats and OGD-treated PC-12/BV2 cells. Downregulation of TRIM27 reduced the brain infarct volume, inflammatory factor levels and brain injury, as well as decreased the number of M1 subtype of microglia cells while increased the number of M2 microglia cells. Moreover, deletion of TRIM27 expression inhibited the expression of p-STAT3, p-NF-κB and HMGB1 in vivo and in vitro. In addition, overexpression of HMGB1 impaired the effects of TRIM27 downregulation on improving OGD-induced cell viability, inhibiting inflammatory reactions and microglia activation. Collectively, this study revealed that TRIM27 was overexpressed in HIE, and downregulation of TRIM27 could alleviate HI-induced brain injury through repressing inflammation and microglia cell activation via the STAT3/HMGB1 axis.
Collapse
Affiliation(s)
- Chengbin Wang
- Department of Pediatrics, Maternal and Child Health Hospital of Xiangyang, Xiangyang, Hubei 441000, China
| | - Pingfeng Zhang
- Department of Pediatrics, Maternal and Child Health Hospital of Xiangyang, Xiangyang, Hubei 441000, China.
| | - Yanni Li
- Department of Pediatrics, Maternal and Child Health Hospital of Xiangyang, Xiangyang, Hubei 441000, China
| | - Xiong Wang
- Department of Pediatrics, Maternal and Child Health Hospital of Xiangyang, Xiangyang, Hubei 441000, China
| | - Lingzhi Guo
- Department of Pediatrics, Maternal and Child Health Hospital of Xiangyang, Xiangyang, Hubei 441000, China
| | - Jingluan Li
- Department of Pediatrics, Maternal and Child Health Hospital of Xiangyang, Xiangyang, Hubei 441000, China
| | - Huihui Jiao
- Department of Pediatrics, Maternal and Child Health Hospital of Xiangyang, Xiangyang, Hubei 441000, China
| |
Collapse
|
22
|
SIRT5 reduces the inflammatory response and barrier dysfunction in IL-17A-induced epidermal keratinocytes. Allergol Immunopathol (Madr) 2023; 51:30-36. [PMID: 36617819 DOI: 10.15586/aei.v51i1.675] [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: 05/18/2022] [Accepted: 06/20/2022] [Indexed: 01/04/2023]
Abstract
Psoriasis is a chronic multisystemic inflammatory disease with inflammatory cell infiltration, hyperproliferation of keratinocytes in skin lesions, and epidermal barrier dysfunction. Normal human epidermal keratinocytes (NHEKs) were stimulated with interleukin 17A (IL-17A). The expression levels of sirtuin-5 (SIRT5) were analyzed by RT-qPCR and western blot assay. The proliferation levels of NHEKs were assessed by EdU staining. The expression of ELOVL1 and ELOVL4 was analyzed by RT-Qpcr, and the expression levels of filaggrin, loricrin, and aquaporin-3 were analyzed by RT-qPCR and western blot. Extracellular signal-regulated kinase 1/2 (ERK1/2) activator t-butylhydroquinone was used to activate ERK1/2. Here, we show that SIRT5 overexpression reduces cell viability and cell proliferation, and improves barrier dysfunction in IL-17A-treated human epidermal keratinocytes, this effect of which is significantly blunted by the ERK1/2 activator. In epidermal keratinocytes, SIRT5 decreases cell proliferation and inflammation and improves barrier dysfunction via ERK/STAT3. This study reveals the role of SIRT5 in the pathogenesis of psoriasis, epidermal hyperplasia, keratinocyte-mediated inflammatory responses, and barrier dysfunction, the role of which is mediated by ERK/STAT3.
Collapse
|
23
|
Sakamoto T, Kuboki S, Furukawa K, Takayashiki T, Takano S, Yoshizumi A, Ohtsuka M. TRIM27-USP7 complex promotes tumour progression via STAT3 activation in human hepatocellular carcinoma. Liver Int 2023; 43:194-207. [PMID: 35753056 DOI: 10.1111/liv.15346] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/31/2022] [Accepted: 06/23/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS TRIM27 is stabilized by binding to USP7 and mediates tumour progression in several cancers; however, the roles of TRIM27-USP7 complex on STAT3 activation in HCC are unknown. METHODS Regulations and functions of TRIM27 for activating STAT3 in HCC were assessed using 207 HCC samples or HCC cells. RESULTS TRIM27 expression was increased in some cases of HCC. High TRIM27 expression was an independent predictor for poor prognosis in HCC after surgery. It was correlated with the expression of EpCAM, vimentin, MMP-9, and activation of STAT3 in HCC. TRIM27 expression was correlated with USP7 expression, and HCC with high TRIM27 expression together with high USP7 expression showed enhanced STAT3 activation, resulting in poorer prognosis. p-JAK1 expression was correlated with STAT3 activation in HCC with high TRIM27 expression. In vitro, USP7 knockdown decreased TRIM27 expression, suggesting that USP7 was essential for TRIM27 stabilization. Knocking down of TRIM27 or USP7 suppressed STAT3 activation and overexpression of TRIM27 accelerated STAT3 activation; therefore, the formation of TRIM27-USP7 complex was needed for STAT3 activation, which led to aggressive tumour proliferation and invasion by enhancing EMT and CSC-like property. Binding of JAK1 to TRIM27-USP7 complex was confirmed in vitro. Deletion of TRIM27-USP7 complex by USP7 inhibitor significantly inhibited tumour cell invasion by suppressing STAT3 activation. CONCLUSIONS TRIM27 is stabilized by binding to USP7 and is related to aggressive tumour progression in HCC via STAT3 activation, resulting in poor prognosis after operation. Therefore, TRIM27-USP7 complex is a useful prognostic predictor and a promising therapeutic target for HCC.
Collapse
Affiliation(s)
- Toshiya Sakamoto
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Kuboki
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Katsunori Furukawa
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tsukasa Takayashiki
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shigetsugu Takano
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Arihito Yoshizumi
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | |
Collapse
|
24
|
Yu C, Rao D, Wang T, Song J, Zhang L, Huang W. Emerging roles of TRIM27 in cancer and other human diseases. Front Cell Dev Biol 2022; 10:1004429. [PMID: 36200036 PMCID: PMC9527303 DOI: 10.3389/fcell.2022.1004429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/05/2022] [Indexed: 12/24/2022] Open
Abstract
As a member of the TRIM protein family, TRIM27 is a RING-mediated E3 ubiquitin ligase that can mark other proteins for degradation. Its ubiquitination targets include PTEN, IκBα and p53, which allows it to regulate many signaling pathways to exert its functions under both physiological and pathological conditions, such as cell proliferation, differentiation and apoptosis. During the past decades, TRIM27 was reported to be involved in many diseases, including cancer, lupus nephritis, ischemia-reperfusion injury and Parkinson’s disease. Although the research interest in TRIM27 is increasing, there are few reviews about the diverse roles of this protein. Here, we systematically review the roles of TRIM27 in cancer and other human diseases. Firstly, we introduce the biological functions of TRIM27. Next, we focus on the roles of TRIM27 in cancer, including ovarian cancer, breast cancer and lung cancer. At the same time, we also describe the roles of TRIM27 in other human diseases, such as lupus nephritis, ischemia-reperfusion injury and Parkinson’s disease. Finally, we discuss the future directions of TRIM27 research, especially its potential roles in tumor immunity.
Collapse
Affiliation(s)
- Chengpeng Yu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Dean Rao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Tiantian Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Song
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jia Song, ; Lei Zhang, ; Wenjie Huang,
| | - Lei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Shanxi Medical University, Jinzhong, China
- Tongji Medical College, Shanxi Tongji Hospital, Huazhong University of Science and Technology, Taiyuan, China
- *Correspondence: Jia Song, ; Lei Zhang, ; Wenjie Huang,
| | - Wenjie Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jia Song, ; Lei Zhang, ; Wenjie Huang,
| |
Collapse
|
25
|
Ni J, Wang P, Yin KJ, Yang XK, Cen H, Sui C, Wu GC, Pan HF. Novel insight into the aetiology of rheumatoid arthritis gained by a cross-tissue transcriptome-wide association study. RMD Open 2022; 8:e002529. [PMID: 37582060 PMCID: PMC9462377 DOI: 10.1136/rmdopen-2022-002529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/23/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Although genome-wide association studies (GWASs) have identified more than 100 loci associated with rheumatoid arthritis (RA) susceptibility, the causal genes and biological mechanisms remain largely unknown. METHODS A cross-tissue transcriptome-wide association study (TWAS) using the unified test for molecular signaturestool was performed to integrate GWAS summary statistics from 58 284 individuals (14 361 RA cases and 43 923 controls) with gene-expression matrix in the Genotype-Tissue Expression project. Subsequently, a single tissue by using FUSION software was conducted to validate the significant associations. We also compared the TWAS with different gene-based methodologies, including Summary Data Based Mendelian Randomization (SMR) and Multimarker Analysis of Genomic Annotation (MAGMA). Further in silico analyses (conditional and joint analysis, differential expression analysis and gene-set enrichment analysis) were used to deepen our understanding of genetic architecture and comorbidity aetiology of RA. RESULTS We identified a total of 47 significant candidate genes for RA in both cross-tissue and single-tissue test after multiple testing correction, of which 40 TWAS-identified genes were verified by SMR or MAGMA. Among them, 13 genes were situated outside of previously reported significant loci by RA GWAS. Both TWAS-based and MAGMA-based enrichment analyses illustrated the shared genetic determinants among autoimmune thyroid disease, asthma, type I diabetes mellitus and RA. CONCLUSION Our study unveils 13 new candidate genes whose predicted expression is associated with risk of RA, providing new insights into the underlying genetic architecture of RA.
Collapse
Affiliation(s)
- Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Peng Wang
- Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, Hefei, China
| | - Kang-Jia Yin
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Xiao-Ke Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Han Cen
- Department of Preventive Medicine, Ningbo University Medical School, Ningbo, Zhejiang, China
| | - Cong Sui
- Department of Orthopedics Trauma, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Guo-Cui Wu
- Department of Obstetrics and Gynecological Nursing, School of Nursing, Anhui Medical University, Hefei, Anhui, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
26
|
Ning M, Liu Y, Wang D, Wei J, Hu G, Xing P. Knockdown of TRIM27 alleviated sepsis-induced inflammation, apoptosis, and oxidative stress via suppressing ubiquitination of PPARγ and reducing NOX4 expression. Inflamm Res 2022; 71:1315-1325. [PMID: 35962797 PMCID: PMC9375190 DOI: 10.1007/s00011-022-01625-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Sepsis is a global fatal disease and leads to severe lung injury due to dysfunction of inflammation response. TRIM27 is closely related to the diseased with dysfunction of inflammation response. The aim of this study was to clarify the role and mechanism of TRIM27 in sepsis-induced lung injury. METHODS The lipopolysaccharide (LPS)-induced septic mouse model was successfully established. The lung injury was evaluated by lung wet/dry (W/D) ratio and hematoxylin-eosin (H&E) staining. The cell apoptosis was evaluated by TUNEL assay. The inflammatory cytokines were measured by quantitative real time-PCR (qRT-PCR) assay and commercial enzyme-linked immunosorbent assay (ELISA). The oxidative stress was assessed by the contents of superoxide dismutase (SOD) and malondialdehyde (MDA), and the expression of dihydroethidium (DHE). RESULTS In this study, we demonstrated that TRIM27 was up-regulated in LPS-induced septic mice. In loss-of-function experiments, knockdown of TRIM27 alleviated sepsis-induced lung injury, inflammation, apoptosis, and oxidative stress. More importantly, knockdown of TRIM27 was observed to reduce p-p65/NOX4 expression via suppressing ubiquitination of PPARγ. In rescue experiments, overexpression of NOX4 abolished the effect of sh-TRIM27 on alleviating sepsis-induced inflammation, apoptosis, and oxidative stress. CONCLUSION These findings highlighted that knockdown of TRIM27 alleviated sepsis-induced inflammation, oxidative stress and apoptosis via suppressing ubiquitination of PPARγ and reducing NOX4 expression, which supports the potential utility of TRIM27 as a therapeutic target in septic lung injury.
Collapse
Affiliation(s)
- Meng Ning
- Department of Heart Center, Tianjin Third Central Hospital, Tianjin, 300170, China.,Department of Heart Center, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, 300170, China.,Department of Heart Center, Artificial Cell Engineering Technology Research Center, Tianjin, 300170, China
| | - Yingwu Liu
- Department of Heart Center, Tianjin Third Central Hospital, Tianjin, 300170, China.,Department of Heart Center, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, 300170, China.,Department of Heart Center, Artificial Cell Engineering Technology Research Center, Tianjin, 300170, China
| | - Donglian Wang
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 222, West Three Road Around Lake, Nanhui New Town, Pudong New Area, Shanghai, China
| | - Jin Wei
- Department of Heart Center, Tianjin Third Central Hospital, Tianjin, 300170, China.,Department of Heart Center, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, 300170, China.,Department of Heart Center, Artificial Cell Engineering Technology Research Center, Tianjin, 300170, China
| | - Guoyong Hu
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 222, West Three Road Around Lake, Nanhui New Town, Pudong New Area, Shanghai, China
| | - Pengcheng Xing
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 222, West Three Road Around Lake, Nanhui New Town, Pudong New Area, Shanghai, China.
| |
Collapse
|
27
|
Tang CH, Qin L, Gao YC, Chen TY, Xu K, Liu T, Ren T. APE1 shRNA-loaded cancer stem cell-derived extracellular vesicles reverse Erlotinib resistance in non-small cell lung cancer via the IL-6/STAT3 signalling. Clin Transl Med 2022; 12:e876. [PMID: 35605028 PMCID: PMC9126360 DOI: 10.1002/ctm2.876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Apurinic endonuclease 1 (APE1) has been suggested as an oncogene of lung tumours and our bioinformatics analysis identified the association between Erlotinib resistance and interleukin-6 (IL-6). Thus, we performed this work to delineate the mechanistic actions of APE1/IL-6 signalling in Erlotinib resistance of non-small cell lung cancer (NSCLC). METHODS We selected human NSCLC cell lines HCC827 and PC9 to establish Erlotinib-resistant HCC827R and PC9R cells. Cancer stem cells (CSCs) were isolated from Erlotinib-sensitive HCC827P and PC9P cells (PCSCs) and from HCC827R and PC9R cells (RCSCs). Further, extracellular vesicles (EVs) were separated from PCSCs (PCSC-EVs) and RCSCs (RCSC-EVs) and co-cultured with RCSCs with or without short hairpin RNA (shRNA)-targeting APE1 (APE1 shRNA) transduction. In addition, functional assays were conducted to determine the effect of APE1 shRNA on malignant phenotypes of cancer cells in vitro and in vivo and the activation of IL-6/STAT3 signalling. RESULTS It was found that NSCLC cells could internalize both RCSC-EVs and PCSC-EVs. RCSC-EVs augmented the resistance of NSCLC cells to Erlotinib. The overexpression of APE1 occurred in NSCLC tissues, and IL-6 was enriched in serum samples of patients with NSCLC. APE1 shRNA was demonstrated to restrict the Erlotinib resistance of NSCLC cells by inactivating the IL-6/STAT3 signalling. Additionally, shAPE1-loaded RCSC-EVs suppressed the Erlotinib resistance of NSCLC via the IL-6/STAT3 axis both in vitro and in vivo, as reflected by impeded malignant phenotypes and xenograft tumour formation. CONCLUSIONS Collectively, these data indicate that APE1 confers Erlotinib resistance by activating the IL-6/STAT3 signalling, suggesting targeting APE1 as a possible therapeutic target in Erlotinib-resistant NSCLC.
Collapse
Affiliation(s)
- Chun-Han Tang
- Clinical Medical College, Chengdu Medical College, Chengdu, P. R. China
| | - Ling Qin
- Department of Gastroenterology, Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, P. R. China
| | - Ying-Chun Gao
- Oncology Department, Pengzhou People's Hospital, Chengdu, P. R. China
| | - Tai-Yu Chen
- Clinical Medical College, Chengdu Medical College, Chengdu, P. R. China
| | - Ke Xu
- Oncology Department, The First Affiliated Hospital of Chengdu Medical College, Chengdu, P. R. China
| | - Tao Liu
- Oncology Department, The First Affiliated Hospital of Chengdu Medical College, Chengdu, P. R. China
| | - Tao Ren
- Clinical Medical College, Chengdu Medical College, Chengdu, P. R. China.,Oncology Department, The First Affiliated Hospital of Chengdu Medical College, Chengdu, P. R. China
| |
Collapse
|
28
|
Ren F, Zhang Y, Qin Y, Shang J, Wang Y, Wei P, Guo J, Jia H, Zhao T. Taraxasterol prompted the anti-tumor effect in mice burden hepatocellular carcinoma by regulating T lymphocytes. Cell Death Dis 2022; 8:264. [PMID: 35577774 PMCID: PMC9110731 DOI: 10.1038/s41420-022-01059-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 01/05/2023]
Abstract
Hepatocellular carcinoma (HCC) is a common digestive malignant tumor with high morbidity and mortality worldwide, however, the treatment of HCC and prognosis of patients are not optimistic, finding more effective treatments are imperative. Taraxacum officinale (L.) Weber ex F.H.Wigg is a perennial herb of compositae, and our study has demonstrated that Taraxacum officinale polysaccharide has certain anti-tumor effect on HCC cells. Taraxasterol (TS) is a natural product extracted from Taraxacum officinale with strong physiological, pharmacological and biological activities, but the effect of TS on HCC is yet to be determined. Therefore, the aim of this study is to explore the effect of dandelion sterol on HCC in vivo and in vitro. The results showed that TS significantly inhibited the proliferation, induced apoptosis and blocked cell cycle in HCC cell lines HepG2 and Huh7 cells in vitro. TS inhibited the tumor growth of H22 bearing mice and the expression of Ki67 in vivo. More importantly, TS regulated the immunity of H22 bearing mice by elevating the ratio of CD4+ T cells in spleen, and increasing the number of T cell infiltration in tumor tissue. Except immunomodulation, the mechanism of tumor growth inhibition may be related to the regulation of apoptosis related proteins and IL-6/STAT3 pathway. TS significantly inhibited the growth of HCC cells both in vitro and in vivo. The study would provide a theoretical basis for the new application of TS and the adjuvant treatment of malignant tumor with traditional Chinese medicine. ![]()
Collapse
Affiliation(s)
- Feng Ren
- Basic Medical College, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China.,Henan International Joint Laboratory of Immunity and Targeted Therapy for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China
| | - Yu Zhang
- Basic Medical College, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China.,Henan International Joint Laboratory of Immunity and Targeted Therapy for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China
| | - Yuanhua Qin
- Basic Medical College, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China.,Henan International Joint Laboratory of Immunity and Targeted Therapy for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China
| | - Jingli Shang
- Basic Medical College, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China.,Henan International Joint Laboratory of Immunity and Targeted Therapy for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China
| | - Yanling Wang
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China.,Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China
| | - Pengkun Wei
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China.,Department of Immunology, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China
| | - Jiaming Guo
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China
| | - Huijie Jia
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China. .,Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China.
| | - Tiesuo Zhao
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China. .,Department of Immunology, Xinxiang Medical University, Xinxiang, 453000, Henan, PR China.
| |
Collapse
|
29
|
Li L, Gao J, Gao L, Li L, Zhang H, Zhao W, Xu S. Bilateral Superior Cervical Sympathectomy Activates Signal Transducer and Activator of Transcription 3 Signal to Alleviate Myocardial Ischemia-Reperfusion Injury. Front Cardiovasc Med 2022; 9:807298. [PMID: 35433880 PMCID: PMC9010611 DOI: 10.3389/fcvm.2022.807298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/10/2022] [Indexed: 11/23/2022] Open
Abstract
Background There is growing evidence about the effect of bilateral superior cervical sympathectomy on myocardial ischemia-reperfusion (I/R) injury. Studies have increasingly found that the signal transducer and activator of transcription 3 (STAT3) plays a protective role in myocardial I/R injury. However, the precise mechanism is unknown. The present study explored the bilateral superior cervical sympathectomy’s effect and potential mechanism in mice myocardial I/R injury. Methods The left heart I/R injury model was created by ligating the anterior descending branch of the coronary artery for 30 min followed by reperfusion. Bilateral superior cervical sympathectomy was performed before myocardial I/R injury. To evaluate the effect of bilateral superior cervical sympathectomy on the myocardium, we examined the myocardial infarct size and cardiac function. Then, myocardial apoptosis, inflammation, and oxidative stress were detected on the myocardium. Furthermore, the expression of STAT3 signal in myocardial tissue was measured by western blotting. To further examine the cardioprotective effect of STAT3 after bilateral superior cervical sympathectomy, the STAT3 inhibitor (static) was utilized to inhibit the phosphorylation of STAT3. Results The results showed that the myocardial I/R injury decreased and the cardiac function recovered in the myocardial I/R injury after cervical sympathectomy. Meanwhile, cervical sympathectomy reduced the myocardial distribution of the sympathetic marker tyrosine hydroxylase (TH) and systemic sympathetic tone. And levels of oxidative stress, inflammatory markers, and apoptosis were reduced in myocardial tissue. We also found that the STAT3 signal was activated in myocardial tissue after cervical sympathectomy. STAT3 inhibitor can partially reverse the myocardial protective effect of cervical sympathectomy. Conclusion Bilateral superior cervical sympathectomy significantly alleviated myocardial I/R injury in mice. And activation of the STAT3 signal may play an essential role in this.
Collapse
|
30
|
Maihofer AX, Choi KW, Coleman JRI, Daskalakis NP, Denckla CA, Ketema E, Morey RA, Polimanti R, Ratanatharathorn A, Torres K, Wingo AP, Zai CC, Aiello AE, Almli LM, Amstadter AB, Andersen SB, Andreassen OA, Arbisi PA, Ashley-Koch AE, Austin SB, Avdibegović E, Borglum AD, Babić D, Bækvad-Hansen M, Baker DG, Beckham JC, Bierut LJ, Bisson JI, Boks MP, Bolger EA, Bradley B, Brashear M, Breen G, Bryant RA, Bustamante AC, Bybjerg-Grauholm J, Calabrese JR, Caldas-de-Almeida JM, Chen CY, Dale AM, Dalvie S, Deckert J, Delahanty DL, Dennis MF, Disner SG, Domschke K, Duncan LE, Džubur Kulenović A, Erbes CR, Evans A, Farrer LA, Feeny NC, Flory JD, Forbes D, Franz CE, Galea S, Garrett ME, Gautam A, Gelaye B, Gelernter J, Geuze E, Gillespie CF, Goçi A, Gordon SD, Guffanti G, Hammamieh R, Hauser MA, Heath AC, Hemmings SMJ, Hougaard DM, Jakovljević M, Jett M, Johnson EO, Jones I, Jovanovic T, Qin XJ, Karstoft KI, Kaufman ML, Kessler RC, Khan A, Kimbrel NA, King AP, Koen N, Kranzler HR, Kremen WS, Lawford BR, Lebois LAM, Lewis C, Liberzon I, Linnstaedt SD, Logue MW, Lori A, Lugonja B, Luykx JJ, Lyons MJ, Maples-Keller JL, Marmar C, Martin NG, Maurer D, Mavissakalian MR, McFarlane A, McGlinchey RE, McLaughlin KA, McLean SA, Mehta D, Mellor R, Michopoulos V, Milberg W, Miller MW, Morris CP, Mors O, Mortensen PB, Nelson EC, Nordentoft M, Norman SB, O'Donnell M, Orcutt HK, Panizzon MS, Peters ES, Peterson AL, Peverill M, Pietrzak RH, Polusny MA, Rice JP, Risbrough VB, Roberts AL, Rothbaum AO, Rothbaum BO, Roy-Byrne P, Ruggiero KJ, Rung A, Rutten BPF, Saccone NL, Sanchez SE, Schijven D, Seedat S, Seligowski AV, Seng JS, Sheerin CM, Silove D, Smith AK, Smoller JW, Sponheim SR, Stein DJ, Stevens JS, Teicher MH, Thompson WK, Trapido E, Uddin M, Ursano RJ, van den Heuvel LL, Van Hooff M, Vermetten E, Vinkers CH, Voisey J, Wang Y, Wang Z, Werge T, Williams MA, Williamson DE, Winternitz S, Wolf C, Wolf EJ, Yehuda R, Young KA, Young RM, Zhao H, Zoellner LA, Haas M, Lasseter H, Provost AC, Salem RM, Sebat J, Shaffer RA, Wu T, Ripke S, Daly MJ, Ressler KJ, Koenen KC, Stein MB, Nievergelt CM. Enhancing Discovery of Genetic Variants for Posttraumatic Stress Disorder Through Integration of Quantitative Phenotypes and Trauma Exposure Information. Biol Psychiatry 2022; 91:626-636. [PMID: 34865855 PMCID: PMC8917986 DOI: 10.1016/j.biopsych.2021.09.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/25/2021] [Accepted: 09/21/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Posttraumatic stress disorder (PTSD) is heritable and a potential consequence of exposure to traumatic stress. Evidence suggests that a quantitative approach to PTSD phenotype measurement and incorporation of lifetime trauma exposure (LTE) information could enhance the discovery power of PTSD genome-wide association studies (GWASs). METHODS A GWAS on PTSD symptoms was performed in 51 cohorts followed by a fixed-effects meta-analysis (N = 182,199 European ancestry participants). A GWAS of LTE burden was performed in the UK Biobank cohort (N = 132,988). Genetic correlations were evaluated with linkage disequilibrium score regression. Multivariate analysis was performed using Multi-Trait Analysis of GWAS. Functional mapping and annotation of leading loci was performed with FUMA. Replication was evaluated using the Million Veteran Program GWAS of PTSD total symptoms. RESULTS GWASs of PTSD symptoms and LTE burden identified 5 and 6 independent genome-wide significant loci, respectively. There was a 72% genetic correlation between PTSD and LTE. PTSD and LTE showed largely similar patterns of genetic correlation with other traits, albeit with some distinctions. Adjusting PTSD for LTE reduced PTSD heritability by 31%. Multivariate analysis of PTSD and LTE increased the effective sample size of the PTSD GWAS by 20% and identified 4 additional loci. Four of these 9 PTSD loci were independently replicated in the Million Veteran Program. CONCLUSIONS Through using a quantitative trait measure of PTSD, we identified novel risk loci not previously identified using prior case-control analyses. PTSD and LTE have a high genetic overlap that can be leveraged to increase discovery power through multivariate methods.
Collapse
Affiliation(s)
- Adam X Maihofer
- Department of Psychiatry, University of California San Diego, La Jolla, California; Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Healthcare System, San Diego, California.
| | - Karmel W Choi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts
| | - Jonathan R I Coleman
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; National Institute of Health Research Maudsley Biomedical Research Centre, King's College London, London, United Kingdom
| | - Nikolaos P Daskalakis
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts; Center of Excellence in Depression and Anxiety Disorders, Belmont, Massachusetts
| | - Christy A Denckla
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Elizabeth Ketema
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Healthcare System, San Diego, California; Research Service, Veterans Affairs Healthcare System, San Diego, California
| | - Rajendra A Morey
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | - Renato Polimanti
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Andrew Ratanatharathorn
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology, Columbia University Mailman School of Public Health, New York
| | - Katy Torres
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Healthcare System, San Diego, California; Research Service, Veterans Affairs Healthcare System, San Diego, California
| | - Aliza P Wingo
- Division of Mental Health, Atlanta Veterans Affairs Medical Center, Decatur, Georgia; Departments of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Clement C Zai
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts; Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Molecular Brain Science, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Allison E Aiello
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Lynn M Almli
- Departments of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Ananda B Amstadter
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Richmond, Virginia
| | - Soren B Andersen
- Research and Knowledge Centre, The Danish Veteran Centre, Ringsted, Denmark
| | - Ole A Andreassen
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Norwegian Centre for Mental Disorders Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Paul A Arbisi
- Mental Health Service, Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota; Department of Psychiatry and Behavioral Sciences, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Allison E Ashley-Koch
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | - S Bryn Austin
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Division of Adolescent and Young Adult Medicine, Boston Children's Hospital, Boston, Massachusetts; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Esmina Avdibegović
- Department of Psychiatry, University Clinical Center of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Anders D Borglum
- Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark; Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
| | - Dragan Babić
- Department of Psychiatry, University Clinical Center of Mostar, Mostar, Bosnia and Herzegovina
| | - Marie Bækvad-Hansen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Dewleen G Baker
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Healthcare System, San Diego, California; Psychiatry Service, Veterans Affairs Healthcare System, San Diego, California
| | - Jean C Beckham
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina; Research Service, Durham Veterans Affairs Medical Center, Durham, North Carolina; Genetics Research Laboratory, Veterans Affairs Mid-Atlantic Mental Illness Research, Education, and Clinical Center, Durham, North Carolina
| | - Laura J Bierut
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, St. Louis, Missouri
| | - Jonathan I Bisson
- Medical Research Council Centre for Psychiatric Genetics and Genomics, National Centre for Mental Health, Cardiff University, Cardiff, United Kingdom
| | - Marco P Boks
- Department of Psychiatry, UMC Utrecht Brain Center, UMC Utrecht, Utrecht, Netherlands
| | - Elizabeth A Bolger
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; McLean Hospital, Belmont, Massachusetts
| | - Bekh Bradley
- Mental Health Services, Decatur, Georgia; Departments of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Meghan Brashear
- Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana
| | - Gerome Breen
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; National Institute of Health Research Maudsley Biomedical Research Centre, King's College London, London, United Kingdom
| | - Richard A Bryant
- Department of Psychology, University of New South Wales, Sydney, New South Wales, Australia
| | - Angela C Bustamante
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jonas Bybjerg-Grauholm
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Joseph R Calabrese
- Department of Psychiatry, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - José M Caldas-de-Almeida
- Lisbon Institute of Global Mental Health, NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal; Chronic Diseases Research Centre, NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal
| | - Chia-Yen Chen
- Translational Biology, Biogen, Cambridge, Massachusetts
| | - Anders M Dale
- Department of Radiology, University of California San Diego, La Jolla, California; Department of Neurosciences, University of California San Diego, La Jolla, California
| | - Shareefa Dalvie
- South African Medical Research Council Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, Cape Town, South Africa; South African Medical Research Council Unit on Child and Adolescent Health, Department of Pediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Jürgen Deckert
- Center of Mental Health, Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany
| | - Douglas L Delahanty
- Department of Psychological Sciences, Kent State University, Kent, Ohio; Research and Sponsored Programs, Kent State University, Kent, Ohio
| | - Michelle F Dennis
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina; Research Service, Durham Veterans Affairs Medical Center, Durham, North Carolina; Genetics Research Laboratory, Veterans Affairs Mid-Atlantic Mental Illness Research, Education, and Clinical Center, Durham, North Carolina
| | - Seth G Disner
- Research Service Line, Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany; Centre for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laramie E Duncan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Alma Džubur Kulenović
- Department of Psychiatry, University Clinical Center of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Christopher R Erbes
- Mental Health Service, Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota; Department of Psychiatry and Behavioral Sciences, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Alexandra Evans
- Medical Research Council Centre for Psychiatric Genetics and Genomics, National Centre for Mental Health, Cardiff University, Cardiff, United Kingdom
| | - Lindsay A Farrer
- Biomedical Genetics Section, Boston University School of Medicine, Boston, Massachusetts; Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts; Department of Epidemiology, Boston University School of Medicine, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Norah C Feeny
- Department of Psychological Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Janine D Flory
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York
| | - David Forbes
- Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia
| | - Carol E Franz
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | - Sandro Galea
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts
| | - Melanie E Garrett
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | - Aarti Gautam
- Center for Military Psychiatry and Neuroscience, Silver Spring, Maryland
| | - Bizu Gelaye
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Joel Gelernter
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut; Departments of Genetics and Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Elbert Geuze
- Department of Psychiatry, UMC Utrecht Brain Center, UMC Utrecht, Utrecht, Netherlands; Brain Research and Innovation Centre, Netherlands Ministry of Defence, Utrecht, Netherlands
| | - Charles F Gillespie
- Departments of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Aferdita Goçi
- Department of Psychiatry, University Clinical Center of Kosovo, Pristina, Kosovo
| | - Scott D Gordon
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Guia Guffanti
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; McLean Hospital, Belmont, Massachusetts
| | - Rasha Hammamieh
- Center for Military Psychiatry and Neuroscience, Silver Spring, Maryland
| | - Michael A Hauser
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Andrew C Heath
- Department of Genetics, Washington University in Saint Louis School of Medicine, St. Louis, Missouri
| | - Sian M J Hemmings
- South African Medical Research Council/Stellenbosch University Extramural Unit on the Genomics of Brain Disorders, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, Cape Town, South Africa; Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, Cape Town, South Africa
| | - David Michael Hougaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Miro Jakovljević
- Department of Psychiatry, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Marti Jett
- Department of Integrative Systems Biology, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Eric Otto Johnson
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, North Carolina; Fellows Program, RTI International, Research Triangle Park, North Carolina
| | - Ian Jones
- Medical Research Council Centre for Psychiatric Genetics and Genomics, National Centre for Mental Health, Cardiff University, Cardiff, United Kingdom
| | - Tanja Jovanovic
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Xue-Jun Qin
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | - Karen-Inge Karstoft
- Research and Knowledge Centre, The Danish Veteran Centre, Ringsted, Denmark; Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Milissa L Kaufman
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; McLean Hospital, Belmont, Massachusetts
| | - Ronald C Kessler
- Department of Health Care Policy, Harvard Medical School, Boston, Massachusetts
| | - Alaptagin Khan
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; McLean Hospital, Belmont, Massachusetts
| | - Nathan A Kimbrel
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina; Mental Health Service, Durham Veterans Affairs Medical Center, Durham, North Carolina; Genetics Research Laboratory, Veterans Affairs Mid-Atlantic Mental Illness Research, Education, and Clinical Center, Durham, North Carolina
| | - Anthony P King
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, Michigan
| | - Nastassja Koen
- South African Medical Research Council Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, Cape Town, South Africa
| | - Henry R Kranzler
- Mental Illness Research, Education and Clinical Center, Corporal Michael J. Crescenz Department of Veterans Affairs Medical Center, Philadelphia, Pennsylvania; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Healthcare System, San Diego, California
| | - Bruce R Lawford
- School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Lauren A M Lebois
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; McLean Hospital, Belmont, Massachusetts
| | - Catrin Lewis
- Medical Research Council Centre for Psychiatric Genetics and Genomics, National Centre for Mental Health, Cardiff University, Cardiff, United Kingdom
| | - Israel Liberzon
- Department of Psychiatry and Behavioral Sciences, Texas A&M University College of Medicine, Bryan, Texas
| | - Sarah D Linnstaedt
- Institute for Trauma Recovery, Department of Anesthesiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mark W Logue
- Biomedical Genetics Section, Boston University School of Medicine, Boston, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts; National Center for PTSD, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Adriana Lori
- Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia
| | - Božo Lugonja
- Medical Research Council Centre for Psychiatric Genetics and Genomics, National Centre for Mental Health, Cardiff University, Cardiff, United Kingdom
| | - Jurjen J Luykx
- Department of Psychiatry, UMC Utrecht Brain Center, UMC Utrecht, Utrecht, Netherlands; Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht, Netherlands
| | - Michael J Lyons
- Dean of Students' Office, Boston University, Boston, Massachusetts
| | - Jessica L Maples-Keller
- Departments of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Charles Marmar
- Department of Psychiatry, New York University School of Medicine, New York
| | - Nicholas G Martin
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Matig R Mavissakalian
- Department of Psychiatry, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Alexander McFarlane
- Centre for Traumatic Stress Studies, University of Adelaide, Adelaide, South Australia, Australia
| | - Regina E McGlinchey
- Translational Research Center for TBI and Stress Disorders, Veterans Affairs Boston Healthcare System, Boston, Massachusetts; Geriatric Research, Education, and Clinical Center, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | | | - Samuel A McLean
- Institute for Trauma Recovery, Department of Anesthesiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Emergency Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Divya Mehta
- School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, Queensland, Australia; Centre for Genomics and Personalised Health, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Rebecca Mellor
- Gallipoli Medical Research Foundation, Greenslopes Private Hospital, Greenslopes, Queensland, Australia
| | - Vasiliki Michopoulos
- Departments of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - William Milberg
- Translational Research Center for TBI and Stress Disorders, Veterans Affairs Boston Healthcare System, Boston, Massachusetts; Geriatric Research, Education, and Clinical Center, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Mark W Miller
- Biomedical Genetics Section, Boston University School of Medicine, Boston, Massachusetts; National Center for PTSD, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Charles Phillip Morris
- School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, Queensland, Australia; Jamieson Trauma Institute, Metro North Hospital and Health Service, Kelvin Grove, Queensland, Australia
| | - Ole Mors
- Psychosis Research Unit, Department of Psychiatry, Aarhus University Hospital, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
| | - Preben B Mortensen
- Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark; Centre for Integrated Register-Based Research, Aarhus University, Aarhus, Denmark; National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
| | - Elliot C Nelson
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, St. Louis, Missouri
| | - Merete Nordentoft
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Mental Health Center Copenhagen, Mental Health Services in the Capital Region of Denmark, University of Copenhagen, Copenhagen, Denmark
| | - Sonya B Norman
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Healthcare System, San Diego, California; Executive Division, National Center for Post-Traumatic Stress Disorder, White River Junction, Vermont
| | - Meaghan O'Donnell
- Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia; Phoenix Australia Centre for Posttraumatic Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Holly K Orcutt
- Department of Psychology, Northern Illinois University, DeKalb, Illinois
| | - Matthew S Panizzon
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | - Edward S Peters
- Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana
| | - Alan L Peterson
- Research and Development Service, South Texas Veterans Health Care System, San Antonio, Texas; Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Matthew Peverill
- Department of Psychology, University of Washington, Seattle, Washington
| | - Robert H Pietrzak
- National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Melissa A Polusny
- Mental Health Service, Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota; Department of Psychiatry and Behavioral Sciences, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - John P Rice
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, St. Louis, Missouri
| | - Victoria B Risbrough
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Healthcare System, San Diego, California; Research Service, Veterans Affairs Healthcare System, San Diego, California
| | - Andrea L Roberts
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Alex O Rothbaum
- Department of Psychological Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Barbara O Rothbaum
- Departments of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Peter Roy-Byrne
- Departments of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington
| | - Kenneth J Ruggiero
- Department of Nursing, Medical University of South Carolina, Charleston, South Carolina; Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Ariane Rung
- Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, Netherlands
| | - Nancy L Saccone
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, St. Louis, Missouri
| | - Sixto E Sanchez
- Department of Medicine, Universidad Peruana de Ciencias Aplicadas Facultad de Ciencias de la Salud, Lima, Peru
| | - Dick Schijven
- Department of Psychiatry, UMC Utrecht Brain Center, UMC Utrecht, Utrecht, Netherlands; Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht, Netherlands
| | - Soraya Seedat
- South African Medical Research Council/Stellenbosch University Extramural Unit on the Genomics of Brain Disorders, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, Cape Town, South Africa; Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, Cape Town, South Africa
| | - Antonia V Seligowski
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; McLean Hospital, Belmont, Massachusetts
| | - Julia S Seng
- Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, Michigan; School of Nursing, University of Michigan, Ann Arbor, Michigan; Department of Women's and Gender Studies, University of Michigan, Ann Arbor, Michigan; Institute for Research on Women and Gender, University of Michigan, Ann Arbor, Michigan
| | - Christina M Sheerin
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Richmond, Virginia
| | - Derrick Silove
- Department of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Alicia K Smith
- Departments of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia; Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia
| | - Jordan W Smoller
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Scott R Sponheim
- Mental Health Service, Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota; Department of Psychiatry and Behavioral Sciences, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Dan J Stein
- South African Medical Research Council Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, Cape Town, South Africa
| | - Jennifer S Stevens
- Departments of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Martin H Teicher
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; Developmental Biopsychiatry Research Program, Belmont, Massachusetts
| | - Wesley K Thompson
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, California; Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Roskilde, Denmark
| | - Edward Trapido
- Department of Epidemiology, School of Public Health, Louisiana State University Health Sciences Center New Orleans, New Orleans, Louisiana
| | - Monica Uddin
- Genomics Program, University of South Florida College of Public Health, Tampa, Florida
| | - Robert J Ursano
- Department of Psychiatry, Uniformed Services University, Bethesda, Maryland
| | - Leigh Luella van den Heuvel
- South African Medical Research Council/Stellenbosch University Extramural Unit on the Genomics of Brain Disorders, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, Cape Town, South Africa; Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, Cape Town, South Africa
| | - Miranda Van Hooff
- Centre for Traumatic Stress Studies, University of Adelaide, Adelaide, South Australia, Australia
| | - Eric Vermetten
- Department of Psychiatry, New York University School of Medicine, New York; Brain Research and Innovation Centre, Netherlands Ministry of Defence, Utrecht, Netherlands; Arq Psychotrauma Research Expert Group, Diemen, Netherlands; Department of Psychiatry, Leiden University Medical Center, Leiden, Netherlands
| | - Christiaan H Vinkers
- Department of Psychiatry, VU University Medical Center Amsterdam, Amsterdam, Netherlands; Department of Anatomy and Neurosciences, VU University Medical Center Amsterdam, Amsterdam, Netherlands
| | - Joanne Voisey
- School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, Queensland, Australia; Centre for Genomics and Personalised Health, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Yunpeng Wang
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark; Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
| | - Zhewu Wang
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina; Department of Mental Health, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Thomas Werge
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
| | - Michelle A Williams
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Douglas E Williamson
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina; Research Service, Durham Veterans Affairs Medical Center, Durham, North Carolina
| | - Sherry Winternitz
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; McLean Hospital, Belmont, Massachusetts
| | - Christiane Wolf
- Center of Mental Health, Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany
| | - Erika J Wolf
- Biomedical Genetics Section, Boston University School of Medicine, Boston, Massachusetts; National Center for PTSD, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Rachel Yehuda
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York; Department of Mental Health, James J. Peters Veterans Affairs Medical Center, Bronx
| | - Keith A Young
- Department of Psychiatry and Behavioral Sciences, Texas A&M University College of Medicine, Bryan, Texas; Department of Psychiatry, Baylor Scott & White Health Central Texas Division, Temple, Texas; Center of Excellence for Research on Returning War Veterans, Central Texas Veterans Health Care System, Waco, Texas
| | - Ross McD Young
- School of Psychology and Counseling, Queensland University of Technology, Kelvin Grove, Queensland, Australia; Jamieson Trauma Institute, Metro North Hospital and Health Service, Kelvin Grove, Queensland, Australia
| | - Hongyu Zhao
- Department of Biostatistics, Yale University, New Haven, Connecticut
| | - Lori A Zoellner
- Departments of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington
| | - Magali Haas
- Cohen Veterans Bioscience, Cambridge, Massachusetts
| | | | | | - Rany M Salem
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California
| | - Jonathan Sebat
- Department of Psychiatry, University of California San Diego, La Jolla, California; Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California; Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Richard A Shaffer
- Department of Epidemiology and Health Sciences, Naval Health Research Center, San Diego, California
| | - Tianying Wu
- Moores Cancer Center, University of California San Diego, La Jolla, California; Division of Epidemiology and Biostatistics, San Diego State University School of Public Health, San Diego, California
| | - Stephan Ripke
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts; Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
| | - Mark J Daly
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; McLean Hospital, Belmont, Massachusetts; Departments of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Karestan C Koenen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Murray B Stein
- Department of Psychiatry, University of California San Diego, La Jolla, California; Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Healthcare System, San Diego, California
| | - Caroline M Nievergelt
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Healthcare System, San Diego, California; Research Service, Veterans Affairs Healthcare System, San Diego, California
| |
Collapse
|
31
|
A systematic comparison of the effect of topically applied anthraquinone aglycones to relieve psoriasiform lesion: The evaluation of percutaneous absorption and anti-inflammatory potency. Biomed Pharmacother 2021; 145:112482. [PMID: 34915669 DOI: 10.1016/j.biopha.2021.112482] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/21/2021] [Accepted: 11/30/2021] [Indexed: 11/21/2022] Open
Abstract
The anthraquinones derived from rhubarb are reported to have anti-inflammatory activity. The present study aimed to assess the topical application of rhubarb anthraquinone aglycones for psoriasis treatment. The antipsoriatic effect of five anthraquinones, including aloe-emodin, rhein, emodin, physcion, and chrysophanol, was compared to elucidate a structure-permeation relationship. Molecular modeling was employed to determine the physicochemical properties. Both macrophages (differentiated THP-1) and keratinocytes (HaCaT) were used to examine the anti-inflammatory activity in the cell-based study. The in vitro pig skin absorption showed that chrysophanol was the compound with the highest cutaneous accumulation. Topically applied rhein was detected to be largely delivered to the receptor compartment. The absorption of rhein was increased by 5-fold in the barrier-deficient skin as compared to intact skin. By stimulating macrophages with imiquimod (IMQ) to model the inflammation in psoriasis, it was found that the anthraquinones significantly reduced IL-6, IL-23, and TNF. The cytokine inhibition level was comparable for the five compounds. The anthraquinones suppressed cytokines by inhibiting the activation of MAPK and NF-κB signaling. The anthraquinones also downregulated IL-6, IL-8, and IL-24 in the inflammatory keratinocytes stimulated with TNF. Rhein and chrysophanol were comparable to curtail the STAT3 phosphorylation in keratinocytes induced by the conditioned medium of stimulated macrophages. The IMQ-induced psoriasiform mouse model demonstrated the improvement of scaling, erythema, and epidermal hyperplasia by topically applied rhein or chrysophanol. The epidermal acanthosis evoked by IMQ was reduced with rhein and chrysophanol by 3-fold. The histological profiles exhibit that both anthraquinone compounds diminished the number of macrophages and neutrophils in the lesional skin, skin-draining lymph node, and spleen. Rhein and chrysophanol showed multifunctional inhibition, by regulating several targets for alleviating psoriasiform inflammation.
Collapse
|
32
|
Antimicrobial and Immunomodulatory Activity of Herb Extracts Used in Burn Wound Healing: "San Huang Powder". EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2900060. [PMID: 34675981 PMCID: PMC8526243 DOI: 10.1155/2021/2900060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/07/2021] [Accepted: 09/11/2021] [Indexed: 12/20/2022]
Abstract
"San Huang Powder," a nonsterile milled herb powder, is frequently used to treat burn wounds in traditional Chinese herbal medicine. However, treating a wound with a nonsterile dressing or reagent is not compatible with the current guidelines in modern medicine. Therefore, we investigated the bactericidal and anti-inflammatory activities of four herb extracts used in "San Huang Powder" in vitro. Meanwhile, an in vivo porcine model with superficial second-degree burns was used for the experiments since the size and skin composition of pigs are the closest to that of the human body. The minimal bactericidal concentration (MBC) of the herb extracts was determined. The in vitro assay indicated that Rhubarb and Phellodendron bark extracts decreased the levels of inflammatory cytokines, IL-8, and GM-CSF on LPS-induced HMEC-1 cells. In accordance with this result, the histopathological evaluation results showed that the efficacy of "San Huang Powder" containing both herb materials was much better than the group without Rhubarb. Our results not only provide a basis to understand why "San Huang Powder" has been used to clinically treat wounds without sterilization directly since ancient times but also show the advantages of using multiple herb materials simultaneously on wound sites to prevent infection during treatment. Rhubarb is the recommended ingredient involved in the preparation of "San Huang Powder" to ensure the healing efficacy of burn wounds.
Collapse
|
33
|
More NB, Sharma N, Pulivendala G, Bale S, Godugu C. Natural product topical therapy in mitigating imiquimod-induced psoriasis-like skin inflammation-underscoring the anti-psoriatic potential of Nimbolide. Indian J Pharmacol 2021; 53:278-285. [PMID: 34414905 PMCID: PMC8411965 DOI: 10.4103/ijp.ijp_591_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Psoriasis is a chronic inflammatory dermatological disorder having complex pathophysiology with autoimmune and genetic factors being the major players. Despite the availability of a gamut of therapeutic strategies, systemic toxicity, poor efficacy, and treatment tolerance due to genetic variability among patients remain the major challenges. This calls for effective intervention with the superior pharmacological profile. Nimbolide (NIM), a major limonoid is an active chemical constituent found in the leaves of the Indian Neem tree, Azadirachta indica. It has gained immense limelight in the past decades for the treatment of various diseases owing to its anti-proliferative, anti-inflammatory, and anti-cancer potentials. OBJECTIVE The present study was centered around evaluating the anti-psoriatic effect of NIM in the experimental model of Imiquimod (IMQ)-induced psoriasis-like inflammation model. MATERIALS AND METHODS Application of IMQ topically on the dorsum of Balb/c mice from day 0-6 prompted psoriasis-like inflammatory symptoms. Treatment groups included topical administration of NIM incorporated carbopol gel formulation and NIM free drug given through subcutaneous route. Protein expression studies such as immunohistochemistry, Western blotting, and ELISA were employed. RESULTS It was clearly observed from our results that NIM significantly ameliorated the expression of inflammatory and proliferation mediators. Further, NIM in the treatment groups significantly improved classic Psoriasis Area Severity Index scoring when compared to IMQ administered group. CONCLUSION It is noteworthy that NIM showed a predominant therapeutic effect as compared to other treatment group. To recapitulate, NIM has shown promising activity as an anti-psoriatic agent by remarkably ameliorating inflammation and associated proliferation.
Collapse
Affiliation(s)
- Nilesh Barku More
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Nivya Sharma
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Gauthami Pulivendala
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Swarna Bale
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| |
Collapse
|
34
|
A Michael Acceptor Analogue, SKSI-0412, Down-Regulates Inflammation and Proliferation Factors through Suppressing Signal Transducer and Activator of Transcription 3 Signaling in IL-17A-Induced Human Keratinocyte. Int J Mol Sci 2021; 22:ijms22168813. [PMID: 34445513 PMCID: PMC8396041 DOI: 10.3390/ijms22168813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 11/17/2022] Open
Abstract
The activation of signal transducer and activator of transcription 3 (STAT3), as well as up-regulation of cytokines and growth factors to promote STAT3 activation, have been found in the epidermis of psoriatic lesions. Recently, a series of synthetic compounds possessing the Michael acceptor have been reported as STAT3 inhibitors by covalently binding to cysteine of STAT3. We synthesized a Michael acceptor analog, SKSI-0412, and confirmed the binding affinity between STAT3 and SKSI-0412. We hypothesized that the SKSI-0412 can inhibit interleukin (IL)-17A-induced inflammation in keratinocytes. The introduction of IL-17A increased the phosphorylation of STAT3 in keratinocytes, whereas the inactivation of STAT3 by SKSI-0412 reduced IL-17A-induced STAT3 phosphorylation and IκBζ expression. In addition, human β defensin-2 and S100A7, which are regulated by IκBζ, were significantly decreased with SKSI-0412 administration. We also confirmed that SKSI-0412 regulates cell proliferation, which is the major phenotype of psoriasis. Based on these results, we suggest targeting STAT3 with SKSI-0412 as a novel therapeutic strategy to regulate IL-17A-induced psoriatic inflammation in keratinocytes.
Collapse
|
35
|
Lv M, Shao J, Jiang F, Liu J. Curcumol may alleviate psoriasis-like inflammation by inhibiting keratinocyte proliferation and inflammatory gene expression via JAK1/STAT3 signaling. Aging (Albany NY) 2021; 13:18392-18403. [PMID: 34314383 PMCID: PMC8351666 DOI: 10.18632/aging.203287] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/14/2021] [Indexed: 12/16/2022]
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by abnormal proliferation and differentiation of keratinocytes. Since curcumol exhibits anti-inflammatory properties in various diseases, we investigated its anti-inflammatory potential in stimulated human keratinocytes. Our data show that curcumol significantly inhibits proliferation and induces cell cycle arrest in NHEK cells stimulated with proinflammatory cytokines (IL-1α, IL-17A, IL-22, oncostatin M, and TNF-α; mix M5). In addition, curcumol markedly ameliorates inflammatory response and promotes differentiation of M5-stimulated NHEK cells. Curcumol inhibits activity of JAK1, resulting in the inhibition of STAT3, downregulation of cyclin D2, and cell cycle arrest in stimulated NHEK cells. Together, our data show that curcumol reduces proliferation and inflammatory gene expression in stimulated keratinocytes by inhibiting the JAK1/STAT3 signaling, suggesting that it might serve as a potential therapeutic option for the treatment of psoriasis.
Collapse
Affiliation(s)
- Mingfen Lv
- Department of Dermatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, P.R. China
| | - Junyi Shao
- Department of Dermatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, P.R. China
| | - Fan Jiang
- Department of Dermatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, P.R. China
| | - Jingjing Liu
- Department of Dermatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, P.R. China
| |
Collapse
|
36
|
Jia X, Zhao C, Zhao W. Emerging Roles of MHC Class I Region-Encoded E3 Ubiquitin Ligases in Innate Immunity. Front Immunol 2021; 12:687102. [PMID: 34177938 PMCID: PMC8222901 DOI: 10.3389/fimmu.2021.687102] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/27/2021] [Indexed: 12/15/2022] Open
Abstract
The major histocompatibility complex (MHC) class I (MHC-I) region contains a multitude of genes relevant to immune response. Multiple E3 ubiquitin ligase genes, including tripartite motif 10 (TRIM10), TRIM15, TRIM26, TRIM27, TRIM31, TRIM38, TRIM39, TRIM40, and RING finger protein 39 (RNF39), are organized in a tight cluster, and an additional two TRIM genes (namely TRIM38 and TRIM27) telomeric of the cluster within the MHC-I region. The E3 ubiquitin ligases encoded by these genes possess important roles in controlling the intensity of innate immune responses. In this review, we discuss the E3 ubiquitin ligases encoded within the MHC-I region, highlight their regulatory roles in innate immunity, and outline their potential functions in infection, inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Xiuzhi Jia
- Department of Pathogenic Biology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunyuan Zhao
- Department of Pathogenic Biology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Pathogenic Biology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
37
|
Chuang SY, Chen CY, Yang SC, Alalaiwe A, Lin CH, Fang JY. 2,4-Dimethoxy-6-Methylbenzene-1,3-diol, a Benzenoid From Antrodia cinnamomea, Mitigates Psoriasiform Inflammation by Suppressing MAPK/NF-κB Phosphorylation and GDAP1L1/Drp1 Translocation. Front Immunol 2021; 12:664425. [PMID: 34054833 PMCID: PMC8162112 DOI: 10.3389/fimmu.2021.664425] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/30/2021] [Indexed: 11/13/2022] Open
Abstract
Antrodia cinnamomea exhibits anti-inflammatory, antioxidant, and immunomodulatory activities. We aimed to explore the antipsoriatic potential of 2,4-dimethoxy-6-methylbenzene-1,3-diol (DMD) derived from A. cinnamomea. The macrophages activated by imiquimod (IMQ) were used as the cell model for examining the anti-inflammatory effect of DMD in vitro. A significantly high inhibition of IL-23 and IL-6 by DMD was observed in THP-1 macrophages and bone marrow-derived mouse macrophages. The conditioned medium of DMD-treated macrophages could reduce neutrophil migration and keratinocyte overproliferation. DMD could downregulate cytokine/chemokine by suppressing the phosphorylation of mitogen-activated protein kinases (MAPKs) and NF-κB. We also observed inhibition of GDAP1L1/Drp1 translocation from the cytoplasm to mitochondria by DMD intervention. Thus, mitochondrial fission could be a novel target for treating psoriatic inflammation. A psoriasiform mouse model treated by IMQ showed reduced scaling, erythema, and skin thickening after topical application of DMD. Compared to the IMQ stimulation only, the active compound decreased epidermal thickness by about 2-fold. DMD diminished the number of infiltrating macrophages and neutrophils and their related cytokine/chemokine production in the lesional skin. Immunostaining of the IMQ-treated skin demonstrated the inhibition of GDAP1LI and phosphorylated Drp1 by DMD. The present study provides insight regarding the potential use of DMD as an effective treatment modality for psoriatic inflammation.
Collapse
Affiliation(s)
- Shih-Yi Chuang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Yuan Chen
- Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shih-Chun Yang
- Department of Cosmetic Science, Providence University, Taichung, Taiwan
| | - Ahmed Alalaiwe
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Chih-Hung Lin
- Center for General Education, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan.,Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
38
|
Han L, Yu R, Ni X, Zhang Z. Effect of microRNA-4268 on Proliferation and Apoptosis of Non-Small Cell Lung Cancer Cells Through Regulating Signal Transducer and Activator of Transcription 3 Level. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
STAT3 is closely related to non-small cell lung cancer (NSCLC). miR-4268 is predicted to regulate STAT3 level by MiRDB analysis. Therefore, our study investigated whether miR-4268 affects NSCLC cells by regulating STAT3. The control group (NC group), miR-4268 Mimics group, and miR-4268
Mimics +pFBD-STAT3 group were set up followed by analysis of miR-4268 and STAT3 mRNA level by QRT-PCR, relationship between miR-4268 and STAT3 by dual fluorescein reporter assay, STAT3 and Tubulin protein level by Western blot, cell proliferation by MTT assay and apoptosis by Annexin V-FITC/PI
staining. Compared with normal tissue, miR-4268 expression in cancer tissue was significantly reduced (P <0.01), while STAT3 level was elevated (P <0.01). STAT3 was a target gene of miR-4268. Compared with NC group, STAT3 level was significantly reduced in miR-4268 Mimics
group (P <0.01) and increased in miR-4268 Mimics+pFBD-STAT3 group compared with miR-4268 Mimics group (P <0.05). Compared to NC group, miR-4268 Mimics group had reduced cell proliferation and increased cell apoptosis and opposite changes were observed in miR-4268 Mimics+pFBD-STAT3
group which had increased cell proliferation and decreased apoptosis (P < 0.05). miR-4268 regulates STAT3 mRNA level and inhibits NSCLC cell proliferation and promotes apoptosis. However, over-expression of STAT3 can inhibit the effect of miR-4268.
Collapse
Affiliation(s)
- Lei Han
- Department of Respiratory, Mudanjiang Medical University Hongqi Hospital, Mudanjiang, Heilongjiang, 157000, China
| | - Renzhi Yu
- Department of Respiratory, Mudanjiang Medical University Hongqi Hospital, Mudanjiang, Heilongjiang, 157000, China
| | - Xin Ni
- Department of Respiratory, Mudanjiang Medical University Hongqi Hospital, Mudanjiang, Heilongjiang, 157000, China
| | - Zenglei Zhang
- Department of Respiratory, Mudanjiang Medical University Hongqi Hospital, Mudanjiang, Heilongjiang, 157000, China
| |
Collapse
|