1
|
Tsoneva Y, Velikova T, Nikolaev G. Circadian clock regulation of myofibroblast fate. Cell Signal 2025; 131:111774. [PMID: 40169063 DOI: 10.1016/j.cellsig.2025.111774] [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: 12/04/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Fibrosis-related disorders represent an increasing medical and economic burden on a worldwide scale, accounting for one-third of all disease-related deaths with limited therapeutic options. As central mediators in fibrosis development, myofibroblasts have been gaining increasing attention in the last 20 years as potential targets for fibrosis attenuation and reversal. While various aspects of myofibroblast physiology have been proposed as treatment targets, many of these approaches have shown limited long-term efficacy so far. However, ongoing research is uncovering new potential strategies for targeting myofibroblast activity, offering hope for more effective treatments in the future. The circadian molecular clock is a feature of almost every cell in the human body that dictates the rhythmic nature of various aspects of human physiology and behavior in response to changes in the surrounding environment. The dysregulation of these rhythms with aging is considered to be one of the underlying reasons behind the development of multiple aging-related chronic disorders, with fibrotic tissue scarring being a common pathological complication among the majority of them. Myofibroblast dysregulation due to skewed circadian clockwork might significantly contribute to fibrotic scar persistence. In the current review, we highlight the role of the circadian clock in the context of myofibroblast activation and deactivation and examine its dysregulation as a driver of fibrogenesis.
Collapse
Affiliation(s)
- Yoanna Tsoneva
- Department of Cell and Developmental Biology, Faculty of Biology, Sofia University "St. Kliment Ohridski", Bulgaria.
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak str, 1407 Sofia, Bulgaria.
| | - Georgi Nikolaev
- Department of Cell and Developmental Biology, Faculty of Biology, Sofia University "St. Kliment Ohridski", Bulgaria.
| |
Collapse
|
2
|
Roztocil E, Husain F, Patrick CC, Feldon SE, Woeller CF. Targeting the Aryl Hydrocarbon Receptor to Attenuate IGF1R Signaling in Thyroid Eye Disease. Thyroid 2025. [PMID: 40257057 DOI: 10.1089/thy.2024.0529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Background: Thyroid eye disease (TED) is an autoimmune disorder characterized by proptosis, inflammation, and fibrosis. Elevated insulin-like growth factor 1 receptor (IGF1R) signaling in TED orbital fibroblasts (OFs) drives the proliferation and biosynthesis of hyaluronan, which causes enlargement of orbital tissue volume. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that regulates cellular stress responses, metabolism, and inflammation. Given its important role in regulating cellular responses, we hypothesized that activation of the AHR could limit excessive IGF1R signaling in TED OFs, offering therapeutic potential. Methods: We measured IGF1R and AHR expression levels in TED, non-TED, and non-OF controls. OF activation was analyzed using proliferation, hyaluronan accumulation, and migration assays. RNA sequencing was used to detect transcriptome-wide changes in IGF1-treated TED OFs. After gene set enrichment analysis, select gene expression changes were validated by quantitative polymerase chain reaction. OFs were treated with the AHR ligands 6-formylindolo[3,2-b]carbazole (FICZ) or tapinarof with or without IGF1. Western blotting evaluated signaling pathways impacted by AHR and IGF1R signaling. Results: TED OFs showed elevated IGF1R and AHR expression levels compared to controls. IGF1 significantly increased hyaluronan accumulation, proliferation, and migration in TED OFs compared to non-TED OFs. IGF1R signaling altered the expression of hundreds of genes controlling cell migration, proliferation, and metabolism in TED OFs. These genes included TUBA1B, TUBA1C, CRABP2 (upregulated), and IRS2 and SOD3 (downregulated). AHR activation blocked proliferation, migration, hyaluronan production, and gene expression mediated through IGF1R signaling. The AHR inhibited these pathways by reducing phosphorylation of GSK3β, an important mediator of IGF1R/β-catenin mediated signaling. Conclusions: AHR activation represents a promising therapeutic strategy for mitigating TED progression by inhibiting IGF1R signaling. Through modulation of GSK3β-mediated pathways, AHR activation may target additional pathologically relevant pathways beyond those affected by direct IGF1R inhibitors. This research provides novel insights into TED pathophysiology and offers a potential avenue for developing therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Elisa Roztocil
- Flaum Eye Institute, University of Rochester, Rochester, New York, USA
| | - Farha Husain
- Flaum Eye Institute, University of Rochester, Rochester, New York, USA
| | | | - Steven E Feldon
- Flaum Eye Institute, University of Rochester, Rochester, New York, USA
| | - Collynn F Woeller
- Flaum Eye Institute, University of Rochester, Rochester, New York, USA
| |
Collapse
|
3
|
Bagheri L, Javanbakht M, Malekian S, Ghahderijani BH, Taghipour S, Tanha FD, Ranjkesh M, Cegolon L, Zhao S. Antifibrotic therapeutic strategies in systemic sclerosis: Critical role of the Wnt/β-catenin and TGF-β signal transduction pathways as potential targets. Eur J Pharmacol 2025:177607. [PMID: 40209848 DOI: 10.1016/j.ejphar.2025.177607] [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: 11/14/2024] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Systemic sclerosis (SSc) is a prototypic fibrosing disorder characterized by widespread fibrosis and immune dysregulation. Current evidence highlights the intricate cross-talk between the canonical Wnt/β-catenin signaling pathway and transforming growth factor-beta (TGF-β) signaling, both of which play fundamental roles in the pathogenesis of fibrosis. This review aims to elucidate the central role of the Wnt/β-catenin-TGF-β pathway and TGF-β signal transduction pathway in fibrotic diseases, focusing on SSc. We summarized evidence from cellular biology studies, animal model investigations, and clinical observations to provide a comprehensive view of the mechanisms by which these pathways cause pathological fibrosis. In addition, we explore the possibilities of antifibrotic therapeutic strategies against Wnt/β-catenin-TGF-β signaling to counteract fibrosis. We aim to delineate approaches towards effectively treating fibrosis in SSc by targeting these interconnected signaling pathways.
Collapse
Affiliation(s)
- Leyla Bagheri
- Department of Internal Medicine, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javanbakht
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sheida Malekian
- Department of Internal Medicine, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sadra Taghipour
- Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Fatemeh Davari Tanha
- Department of Infertility, Yas Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Luca Cegolon
- Department of Medical, Surgical & Health Sciences, University of Trieste, 34128 Trieste, Italy; Public Health Department, University Health Agency Giuliano-Isontina (ASUGI), 34148 Trieste, Italy
| | - Shi Zhao
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
4
|
Xue C, Chu Q, Shi Q, Zeng Y, Lu J, Li L. Wnt signaling pathways in biology and disease: mechanisms and therapeutic advances. Signal Transduct Target Ther 2025; 10:106. [PMID: 40180907 PMCID: PMC11968978 DOI: 10.1038/s41392-025-02142-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/13/2024] [Accepted: 12/29/2024] [Indexed: 04/05/2025] Open
Abstract
The Wnt signaling pathway is critically involved in orchestrating cellular functions such as proliferation, migration, survival, and cell fate determination during development. Given its pivotal role in cellular communication, aberrant Wnt signaling has been extensively linked to the pathogenesis of various diseases. This review offers an in-depth analysis of the Wnt pathway, detailing its signal transduction mechanisms and principal components. Furthermore, the complex network of interactions between Wnt cascades and other key signaling pathways, such as Notch, Hedgehog, TGF-β, FGF, and NF-κB, is explored. Genetic mutations affecting the Wnt pathway play a pivotal role in disease progression, with particular emphasis on Wnt signaling's involvement in cancer stem cell biology and the tumor microenvironment. Additionally, this review underscores the diverse mechanisms through which Wnt signaling contributes to diseases such as cardiovascular conditions, neurodegenerative disorders, metabolic syndromes, autoimmune diseases, and cancer. Finally, a comprehensive overview of the therapeutic progress targeting Wnt signaling was given, and the latest progress in disease treatment targeting key components of the Wnt signaling pathway was summarized in detail, including Wnt ligands/receptors, β-catenin destruction complexes, and β-catenin/TCF transcription complexes. The development of small molecule inhibitors, monoclonal antibodies, and combination therapy strategies was emphasized, while the current potential therapeutic challenges were summarized. This aims to enhance the current understanding of this key pathway.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
5
|
Bhatt J, Ghigo A, Hirsch E. PI3K/Akt in IPF: untangling fibrosis and charting therapies. Front Immunol 2025; 16:1549277. [PMID: 40248697 PMCID: PMC12004373 DOI: 10.3389/fimmu.2025.1549277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/13/2025] [Indexed: 04/19/2025] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a chronic, progressive lung disease characterized by abnormal epithelial repair, persistent inflammation, and excessive extracellular matrix deposition, leading to irreversible scarring and respiratory failure. Central to its pathogenesis is the dysregulation of the PI3K/Akt signaling pathway, which drives fibroblast activation, epithelial-mesenchymal transition, apoptosis resistance, and cellular senescence. Senescent cells contribute to fibrosis through the secretion of pro-inflammatory and profibrotic factors in the senescence-associated secretory phenotype (SASP). Current antifibrotic therapies, Nintedanib and Pirfenidone, only slow disease progression and are limited by side effects, highlighting the need for novel treatments. This review focuses on the role of PI3K/Akt signaling in IPF pathogenesis, its intersection with inflammation and fibrosis, and emerging therapeutic approaches targeting molecules along this pathway.
Collapse
Affiliation(s)
- Janki Bhatt
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| |
Collapse
|
6
|
Patil VS, Patil CR, Patel HM, Kumar A. Exploring disulfiram mechanisms in renal fibrosis: insights from biological data and computational approaches. Front Pharmacol 2025; 16:1480732. [PMID: 40170735 PMCID: PMC11958968 DOI: 10.3389/fphar.2025.1480732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/03/2025] [Indexed: 04/03/2025] Open
Abstract
Background Disulfiram (DSF) is an anti-alcoholic drug that has been reported to inhibit the epithelial-to-mesenchymal transition and crosslinking during fibrosis, pyroptosis, and inflammatory NF-κB and Nrf-2 signaling pathways. However, there is insufficient evidence to support the mechanisms of DSF in preventing renal fibrosis (RF). Therefore, the current study aimed to elucidate the DSF-modulated targets and pathways in renal fibrosis. Methods The common proteins between DSF and RF were screened for protein-protein interaction, pathway enrichment, cluster, and gene ontology analysis. Molecular docking was executed for core genes using AutoDock Vina through the POAP pipeline. Molecular dynamics (MD) simulation (100 ns) was performed to infer protein-ligand stability, and conformational changes were analyzed by free energy landscape (FEL). Results A total of 78 targets were found to be common between DSF and RF, of which NFKB, PIK3CA/R1, MTOR, PTGS2, and MMP9 were the core genes. PI3K-Akt signaling followed by JAK-STAT, TNF, Ras, ErbB, p53, phospholipase D, mTOR, IL-17, NF-κB, AMPK, VEGF, and MAPK signaling pathways were modulated by DSF in RF. DSF showed a direct binding affinity with active site residues of core genes, and except for DSF with NF-κB, all other complexes, including the standard, were found to be stable during 100 ns MD simulation with minimal protein-ligand root mean squared deviation and residual fluctuations and higher compactness with broad conformational changes. Conclusion DSF protects against renal fibrosis, and this study paves the way for experimental investigation to repurpose DSF for treating RF.
Collapse
Affiliation(s)
- Vishal S. Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Chandragouda R. Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Harun M. Patel
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Anoop Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| |
Collapse
|
7
|
Huang L, Chen Y, Fan X, Zhang X, Wang X, Liu L, Liu T, Wang P, Xu A, Zhao X, Cong M. Fluorofenidone mitigates liver fibrosis through GSK-3β modulation and hepatocyte protection in a 3D tissue-engineered model. Int Immunopharmacol 2025; 149:114209. [PMID: 39919455 DOI: 10.1016/j.intimp.2025.114209] [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: 01/01/2025] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/09/2025]
Abstract
Liver fibrosis, a critical stage in chronic liver disease progression, presents a significant global health challenge. This study investigates the antifibrotic and hepatoprotective properties of fluorofenidone (AKF-PD) using a 3D tissue-engineered model. A 3D in vitro liver fibrosis model was developed using decellularized rat liver scaffolds seeded with hepatocytes, hepatic stellate cells (HSCs), and sinusoidal endothelial cells to replicate the multicellular liver microenvironment. The model was stimulated with carbon tetrachloride (CCl4) to induce fibrotic conditions, resulting in collagen deposition, HSC activation, and elevated fibrosis markers. Parallel in vivo studies employed C57BL/6J mice with CCl4-induced liver fibrosis. The antifibrotic and hepatoprotective effects of AKF-PD were evaluated by assessing collagen deposition, fibrosis markers, and hepatocyte apoptosis. Oxidative stress markers and inflammation-related proteins were also measured. Molecular docking identified GSK-3β as a target protein of AKF-PD, and subsequent analyses explored the GSK-3β/β-catenin and Nrf2/HO-1 signaling pathways. AKF-PD demonstrated significant efficacy in reducing fibrosis markers and protecting hepatocytes by inhibiting apoptosis and oxidative stress. Mechanistically, AKF-PD targets the GSK-3β/β-catenin pathway, suppressing β-catenin-mediated pro-fibrotic gene expression, while activating the Nrf2/HO-1 pathway to mitigate oxidative stress, thereby reducing hepatocyte apoptosis. These findings are consistent with results from CCl4-induced mouse fibrosis models, validating the 3D model's applicability for preclinical drug evaluation. This 3D liver fibrosis model provides a physiologically relevant platform for studying fibrosis and anti-fibrotic mechanisms, highlighting AKF-PD's promise as a therapeutic agent and advancing liver fibrosis research.
Collapse
Affiliation(s)
- Long Huang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University,Beijing, China; State Key Laboratory of Digestive Health and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Yu Chen
- Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University; Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, China
| | - Xu Fan
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University,Beijing, China; State Key Laboratory of Digestive Health and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Xiaohui Zhang
- Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University; Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, China
| | - Xue Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University,Beijing, China; State Key Laboratory of Digestive Health and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Lin Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University,Beijing, China; State Key Laboratory of Digestive Health and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Tianhui Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University,Beijing, China; State Key Laboratory of Digestive Health and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University,Beijing, China; State Key Laboratory of Digestive Health and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Anjian Xu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University,Beijing, China; State Key Laboratory of Digestive Health and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University,Beijing, China; State Key Laboratory of Digestive Health and National Clinical Research Center of Digestive Disease, Beijing, China.
| |
Collapse
|
8
|
Chen L, Yang H, Wang J, Zhang H, Fu K, Yan Y, Liu Z. MEOX1-mediated transcriptional regulation of circABHD3 exacerbates hepatic fibrosis through promoting m6A/YTHDF2-dependent YPEL3 mRNA decay to activate β-catenin signaling. PLoS Genet 2025; 21:e1011622. [PMID: 40100806 PMCID: PMC11918346 DOI: 10.1371/journal.pgen.1011622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Hepatic fibrosis may progress to liver cirrhosis and eventually cause death. Epithelial-mesenchymal transition (EMT) of hepatocytes plays critical roles in hepatic fibrosis. Exploring the mechanisms underlying EMT is crucial for a better understanding of hepatic fibrosis pathogenesis. METHODS Hepatocyte EMT wad induced with TGF-β1 and evaluated by Western blotting and immunofluorescence staining. Methylated RNA immunoprecipitation (MeRIP) was applied to assess N6-methyladenosine (m6A) modification. RIP and RNA pull-down assays were performed to analyze the interaction between circABHD3, YTHDF2 and YPEL3 mRNA. MEOX1-mediated transcription of ABHD3 was examined by luciferase and chromatin immunoprecipitation (ChIP). Mice were intraperitoneally injected with CCl4 or treated with bile duct ligation (BDL) surgery for hepatic fibrosis induction. Liver injury and collagen deposition were examined with hematoxylin and eosin (HE), Masson, and Sirius Red staining. Alanine transaminase (ALT), aspartate transaminase (AST) and hydroxyproline (HYP) were examined using ELISA. RESULTS CircABHD3 was upregulated in in vitro and in vivo models of hepatic fibrosis and patients. Knockdown of circABHD3 inhibited TGF-β1-induced expression of fibrosis markers, EMT and mitochondrial impairment in hepatocytes. MEOX1 could directly bind to the promoter of ABHD3 to facilitate its transcription and subsequent circABHD3 generation. Knockdown of MEOX1 suppressed TGF-β1-induced EMT and mitochondrial impairment through suppression of circABHD3. CircABHD3 destabilized YPEL3 mRNA via promoting YTHDF2-dependent recognition of m6A-modified YPEL3 mRNA to trigger β-catenin signaling activation. Furthermore, circABHD3 silencing-mediated inhibition of EMT and mitochondrial impairment was counteracted by YPEL3 knockdown and activation of β-catenin signaling. Depletion of circABHD3 significantly reduced EMT, mitochondrial impairment and hepatic fibrosis via promoting YPEL3 expression and suppressing β-catenin signaling in vivo. CONCLUSION MEOX1-mediated generation of circABHD3 promotes EMT and mitochondrial impairment by enhancing YTHDF2-mediated degradation of YPEL3 mRNA and activating downstream β-catenin signaling, thus exacerbating hepatic fibrosis.
Collapse
Affiliation(s)
- Limin Chen
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Hui Yang
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Juan Wang
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Haoye Zhang
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Kangkang Fu
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Yu Yan
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Zhenguo Liu
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| |
Collapse
|
9
|
Sabini JH, Timotius KH. Hepatoprotective and Fat-Accumulation-Reductive Effects of Curcumin on Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Curr Issues Mol Biol 2025; 47:159. [PMID: 40136412 PMCID: PMC11940900 DOI: 10.3390/cimb47030159] [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: 01/16/2025] [Revised: 02/06/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Fat accumulation is the hallmark of metabolic dysfunction-associated steatotic liver disease (MASLD). Given the intimidating nature of its treatment, curcumin (CUR) emerges as a potential therapeutic agent due to its proven effectiveness in managing MASLD. This review aimed to evaluate previous reports on the hepatoprotective and fat-accumulation-reductive effects of CUR administration in preventing or treating MASLD. CUR administration can modulate serum liver enzymes and lipid profiles. The fat accumulation of MASLD is the primary cause of oxidative stress and inflammation. By reducing fat accumulation, CUR may attenuate the inflammation and oxidative stress in MASLD. In addition, CUR has been proven to restore the dysfunctional cellular energy metabolism capacity and attenuate fibrogenesis (antifibrotic agent). Their hepatoprotective effects are associated with fat accumulation in MASLD. Lipid metabolism (lipogenesis, lipolysis, and lipophagy) is correlated with their hepatoprotective effects. CUR has prophylactic and therapeutic effects, particularly in early-stage MASLD, primarily when it is used as a fat reducer. It can be considered an excellent natural therapeutic drug for MASLD because it protects the liver and attenuates fat accumulation, especially in the early stage of MASLD development.
Collapse
Affiliation(s)
| | - Kris Herawan Timotius
- Faculty of Medicine and Health Sciences, Krida Wacana Christian University, Jakarta 11510, Indonesia;
| |
Collapse
|
10
|
Han J, Wang J, Shen L, Cai Y, Wang X, Wumaier A, Chen W, Han W. Aging-enhanced autophagy activity promotes fibrotic progression via the TGF-β2/Smad signaling pathway in trabecular meshwork cells-a new insight from POAG. Front Med (Lausanne) 2025; 11:1534120. [PMID: 39882536 PMCID: PMC11774994 DOI: 10.3389/fmed.2024.1534120] [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: 11/25/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Glaucoma, a leading cause of irreversible blindness, is characterized by optic neuropathy and retinopathy, with primary open-angle glaucoma (POAG) being the most prevalent form. The primary pathogenic mechanism of POAG involves elevated intraocular pressure caused by chronic fibrosis of the trabecular meshwork (TM). Autophagy, a critical process for maintaining cellular homeostasis, has been implicated in fibrosis across various organs. However, its precise role in the fibrosis associated with POAG pathogenesis remains unclear. This study investigates the involvement of autophagy in TM fibrosis and explores its potential impact on POAG development, aiming to provide insights into new therapeutic targets. Methods To assess autophagy activity and its relationship with fibrosis, we analyzed TM tissues from POAG patients and healthy donors. Autophagic activity in human TM tissues was measured through immunohistochemical analyses. An in vitro aging model using chronic H2O2 treatment was established to investigate the change of fibrosis in TM cells. Additionally, we used dexamethasone-treated TM cells as a POAG model to explore the role of autophagy in fibrotic progression. The involvement of the TGF-β2/Smad signaling pathway was investigated through western blot analysis and quantitative real-time PCR. Results This study reveals increased autophagic activity in tissues from POAG patients and an age-related upregulation of autophagy in healthy human TM tissues. In the H2O2-induced aging model, TM cells displayed both elevated autophagic activity and fibrosis. Further investigation showed that enhanced autophagy activity promoted fibrotic progression via activation of the TGF-β2/Smad signaling pathway. Similarly, in the dexamethasone-treated TM cell model, autophagy was found to exacerbate fibrosis, aligning with observations in the aging model. Discussion In this study, we uncover the interplay between autophagy and the TGF-β2/Smad pathway in the pathogenesis of POAG. We observed increased autophagic activity in TM tissues from POAG patients and in TM tissues of aging healthy individuals. In human primary TM cells, we confirmed that autophagy becomes activated in the context of cellular senescence and the development of POAG, which further facilitates fibrotic progression via the TGF-β2/Smad signaling pathway. These findings underscore the important role of autophagy in POAG pathogenesis and confirm senescence as a pivotal risk factor.
Collapse
Affiliation(s)
- Jin Han
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jun Wang
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ling Shen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yiting Cai
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xuze Wang
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ailixiati Wumaier
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei Chen
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei Han
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Alotaiq N, Khalifa AS, Youssef A, El-Nagar EG, Elwali NE, Habib HM, AlZaim I, Eid AH, Bakkar NMZ, El-Yazbi AF. Targeting GSK-3β for adipose dysfunction and cardiovascular complications of metabolic disease: An entangled WNT/β-catenin question. FASEB J 2024; 38:e70273. [PMID: 39726401 DOI: 10.1096/fj.202402470r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Individuals with metabolic syndrome have a high risk of developing cardiovascular disorders that is closely tied to visceral adipose tissue dysfunction, as well as an altered interaction between adipose tissue and the cardiovascular system. In metabolic syndrome, adipose tissue dysfunction is associated with increased hypertrophy, reduced vascularization, and hypoxia of adipocytes, leading to a pro-oxidative and pro-inflammatory environment. Among the pathways regulating adipose tissue homeostasis is the wingless-type mammary tumor virus integration site family (Wnt) signaling pathway, with both its canonical and non-canonical arms. Various modulators of the Wnt signaling have been identified to contribute to the development of metabolic diseases and their cardiovascular complications, with a particularly significant role played by Glycogen Synthase Kinase-3β (GSK-3β). GSK-3β levels and activities have various and often contrasting roles in obesity and related metabolic disorders, as well as their cardiovascular sequelae. Here, we explore the possibility that altered Wnt signaling and GSK-3β activities could serve as a connection between adipose tissue dysfunction and the development of cardiovascular disease in individuals with metabolic syndrome. We attempt to define a context-specific approach for intervention, which could possibly serve as a novel disease modifying therapy for the mitigation of such complications.
Collapse
Affiliation(s)
- Nasser Alotaiq
- Health Sciences Research Center, Imam Muhammad Ibn Saud Islamic University (IMISIU), Riyadh, Kingdom of Saudi Arabia
| | - Ahmed S Khalifa
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Amr Youssef
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Esraa G El-Nagar
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Nasr Eldin Elwali
- Deanship of Scientific Research, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Kingdom of Saudi Arabia
| | - Hosam M Habib
- Research & Innovation Hub, Alamein International University, Alamein, Egypt
| | - Ibrahim AlZaim
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Ahmed F El-Yazbi
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
- Research & Innovation Hub, Alamein International University, Alamein, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
12
|
Tan X, Cao M, Zhao Y, Yi L, Li Y, He C, Li QX, Dong Y. Neuroprotection of isoorientin against microglia activation induced by lipopolysaccharide via regulating GSK3β, NF-κb and Nrf2/HO-1 pathways. Immunopharmacol Immunotoxicol 2024; 46:741-750. [PMID: 39245870 DOI: 10.1080/08923973.2024.2399249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
Background: Isoorientin (ISO), a flavone C-glycoside, is a glycogen synthase kinase 3β (GSK3β) substrate-competitive inhibitor. ISO has potential in treatment of Alzheimer's disease (AD). An excessive activation of GSK3β can lead to neuroinflammation causing neuronal damage. Microglia cells, as resident immune cells of the central nervous system, mediate neuroinflammation. Here, we studied the effects of ISO on microglial activation to alleviate neuroinflammation. Methods: Effects of ISO were observed upon the stimulation of mouse microglia BV2 or SIM-A9 cells by lipopolysaccharide (LPS). Lithium chloride (LiCl) was the positive control as a GSK3β inhibitor. The release of TNF-α and NO were analyzed by ELISA and Griess assays, while expressions of COX-2, Iba-1, BDNF, GSK3β, NF-κB p65, IκB, Nrf2 and HO-1 were detected by Western blotting. In the co-culture model of SIM-A9 cells and differentiated SH-SY5Y human neuroblastoma cells, effects of ISO on microglia-mediated neuronal damage were evaluated with the MTS assay. Results: ISO significantly inhibited the production of TNF-α (p < 0.01), NO (p < 0.001) and the expression of COX-2 (p < 0.01) and Iba-1 (p < 0.05) induced by LPS, and increased BDNF. The cell viability of SH-SY5Y was inhibited by LPS in the co-culture, which was prevented by ISO pretreatment. ISO increased the expression of p-GSK3β (Ser9), IκB and HO-1 in the cytoplasm, decreased NF-κB p65 and increased Nrf2 in the nucleus compared with the LPS group. Conclusion: ISO attenuated the activation of microglia through regulating the GSK3β, NF-κB and Nrf2/HO-1 signaling pathways to exert neuroprotection.
Collapse
Affiliation(s)
- Xiaoqin Tan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, Hawaii, USA
- Medical Department, Wuhan City Collge, Wuhan, China
| | - Mindie Cao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Yijing Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Lang Yi
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Yingui Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Changhong He
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Qing X Li
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Yan Dong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| |
Collapse
|
13
|
Zhang H, Li C, Li W, Xin W, Qin T. Research Advances in Adenomyosis-Related Signaling Pathways and Promising Targets. Biomolecules 2024; 14:1402. [PMID: 39595579 PMCID: PMC11591984 DOI: 10.3390/biom14111402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/26/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Adenomyosis is a benign gynecological condition characterized by the proliferation of the endometrial stroma and glands into the myometrium, uterine volume enlargement, and peripheral smooth muscle hypertrophy. The typical clinical symptoms include chronic pelvic pain, abnormal uterine bleeding, and subfertility, all of which significantly impact quality of life. There are no effective prevention or treatment strategies for adenomyosis, partly due to a limited understanding of the pathological mechanisms underlying the initiation and progression of the disease. Given that signaling pathways play a crucial role in the development of adenomyosis, a better understanding of these signaling pathways is essential for identifying therapeutic targets and advancing drug development. The occurrence and progression of adenomyosis are closely linked to various underlying pathophysiological mechanisms, including proliferation, migration, invasion, fibrosis, angiogenesis, inflammation, oxidative stress, immune response, and epigenetic changes. This review summarizes the signaling pathways and targets associated with the pathogenesis of adenomyosis, including CXCL/CXCR, NLRP3, NF-κB, TGF-β/smad, VEGF, Hippo/YAP, PI3K/Akt/mTOR, JAK/STAT, and other relevant pathways. In addition, it identifies promising future targets for the development of adenomyosis treatment, such as m6A, GSK3β, sphks, etc.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Gynecology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Chaoming Li
- Departmemt of Urology, The First People’s Hospital of Longnan, Longnan 742500, China
| | - Wenyan Li
- Department of Gynecology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Wenhu Xin
- Department of Gynecology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Tiansheng Qin
- Department of Gynecology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
14
|
Palanisamy K, Karpagavalli M, Nareshkumar RN, Ramasubramanyan S, Angayarkanni N, Raman R, Chidambaram S. Adiponectin-induced activation of ERK1/2 drives fibrosis in retinal pigment epithelial cells. Hum Cell 2024; 38:8. [PMID: 39460900 DOI: 10.1007/s13577-024-01131-5] [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: 08/26/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024]
Abstract
Adiponectin (APN), a vasoactive cytokine produced by adipocytes, has emerged as a critical player in retinal diseases. Renowned for its antioxidant, anti-angiogenic, and anti-inflammatory properties, APN levels are closely linked to metabolic disorders, such as insulin resistance, obesity, and diabetic retinopathy (DR). Our previous work demonstrated that APN is similar in efficiency as Avastin in limiting neovascularization in retinal endothelial cells. In this study, we analyzed the effect of APN on retinal epithelial cells to understand its potential impact on eye-related pathologies. Overexpression of APN in ARPE-19 cells predominantly yielded the MMW-APN form, accompanied by increased expression of pro-fibrotic markers and decreased levels of tight junction (TJ) proteins, ZO-1, and Occludin. Further, confocal imaging revealed impaired TJ assembly and the integrity of TJ was also compromised as evidenced by the higher paracellular permeability and lower TEER. Besides, rAPN treatment in ARPE-19 cells as well triggered increased expression of pro-fibrotic markers, pro-MMP2, and enhanced cell migration and proliferation. Mechanistically, these pro-fibrotic effects were mediated by APN-induced phosphorylation of ERK1/2, causing RPE cell transdifferentiation. Furthermore, we identified that MMW-APN was the most prevalent form detected in the vitreous humor of proliferative diabetic retinopathy (PDR) patients, emphasizing the clinical relevance of our findings. Overall, our data suggest that APN, particularly its MMW form, induces epithelial-mesenchymal transition (EMT) and fibrosis in RPE cells, potentially driving the angio-fibrotic shift observed in PDR via ERK1/2 activation.
Collapse
Affiliation(s)
- Karthikka Palanisamy
- Department of Biochemistry and Cell Biology, R.S. Mehta Jain, KBIRVO, Vision Research Foundation, Chennai, India
| | | | | | - Sharada Ramasubramanyan
- Department of Biochemistry and Cell Biology, R.S. Mehta Jain, KBIRVO, Vision Research Foundation, Chennai, India
| | - Narayanasamy Angayarkanni
- Department of Biochemistry and Cell Biology, R.S. Mehta Jain, KBIRVO, Vision Research Foundation, Chennai, India
| | - Rajiv Raman
- Shri Bhagwan Mahavir Vitreoretinal Services, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | | |
Collapse
|
15
|
Chen W, Gao Y, Liu Y, Luo Y, Xue X, Xiao C, Wei K. Tanshinone IIA Loaded Inhaled Polymer Nanoparticles Alleviate Established Pulmonary Fibrosis. ACS Biomater Sci Eng 2024; 10:6250-6262. [PMID: 39288315 DOI: 10.1021/acsbiomaterials.4c00532] [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: 09/19/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal respiratory disease characterized by chronic, progressive scarring of the lung parenchyma, leading to an irreversible decline in lung function. Apart from supportive care, there is currently no specific treatment available to reverse the disease. Based on the fact that tanshinone IIA (TAN) had an effect on protecting against TGF-β1-induced fibrosis through the inhibition of Smad and non-Smad signal pathways to avoid myofibroblasts activation, this study reported the development of the inhalable tanshinone IIA-loaded chitosan-oligosaccharides-coated poly(lactic-co-glycolic acid) (PLGA) nanoparticles (CPN@TAN) for enhancing the pulmonary delivery of tanshinone IIA to treat pulmonary fibrosis. The CPN@TAN with a size of 206.5 nm exhibited excellent in vitro aerosol delivery characteristics, featuring a mass median aerodynamic diameter (MMAD) of 3.967 ± 0.025 μm and a fine particle fraction (FPF) of 70.516 ± 0.929%. Moreover, the nanoparticles showed good stability during atomization and enhanced the mucosal penetration capabilities. The results of confocal spectroscopy confirmed the potential of the nanoparticles as carriers that facilitated the uptake of drugs by NIH3T3, A549, and MH-S cells. Additionally, the nanoparticles demonstrated good in vitro biocompatibility. In a mouse model of bleomycin-induced pulmonary fibrosis, noninvasive inhalation of aerosol CPN@TAN greatly suppressed collagen formation and facilitated re-epithelialization of the destroyed alveolar epithelium without causing systemic toxicity compared with intravenous administration. Consequently, our noninvasive inhalation drug delivery technology based on polymers may represent a promising paradigm and open the door to overcoming the difficulties associated with managing pulmonary fibrosis.
Collapse
Affiliation(s)
- Wenyu Chen
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yuanyuan Gao
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yuanqi Liu
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yujia Luo
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Xinrui Xue
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Chujie Xiao
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Kun Wei
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
16
|
Haque M, Shyanti RK, Mishra MK. Targeted therapy approaches for epithelial-mesenchymal transition in triple negative breast cancer. Front Oncol 2024; 14:1431418. [PMID: 39450256 PMCID: PMC11499239 DOI: 10.3389/fonc.2024.1431418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is distinguished by negative expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), making it an aggressive subtype of breast cancer and contributes to 15-20% of the total incidence. TNBC is a diverse disease with various genetic variations and molecular subtypes. The tumor microenvironment involves multiple cells, including immune cells, fibroblast cells, extracellular matrix (ECM), and blood vessels that constantly interact with tumor cells and influence each other. The ECM undergoes significant structural changes, leading to induced cell proliferation, migration, adhesion, invasion, and epithelial-to-mesenchymal transition (EMT). The involvement of EMT in the occurrence and development of tumors through invasion and metastasis in TNBC has been a matter of concern. Therefore, EMT markers could be prognostic predictors and potential therapeutic targets in TNBC. Chemotherapy has been one of the primary options for treating patients with TNBC, but its efficacy against TNBC is still limited. Targeted therapy is a critical emerging option with enhanced efficacy and less adverse effects on patients. Various targeted therapy approaches have been developed based on the specific molecules and the signaling pathways involved in TNBC. These include inhibitors of signaling pathways such as TGF-β, Wnt/β-catenin, Notch, TNF-α/NF-κB and EGFR, as well as immune checkpoint inhibitors, such as pembrolizumab, 2laparib, and talazoparib have been widely explored. This article reviews recent developments in EMT in TNBC invasion and metastasis and potential targeted therapy strategies.
Collapse
Affiliation(s)
| | | | - Manoj K. Mishra
- Cancer Research Center, Department of Biological Sciences, Alabama State
University, Montgomery, AL, United States
| |
Collapse
|
17
|
Kühl F, Brand K, Lichtinghagen R, Huber R. GSK3-Driven Modulation of Inflammation and Tissue Integrity in the Animal Model. Int J Mol Sci 2024; 25:8263. [PMID: 39125833 PMCID: PMC11312333 DOI: 10.3390/ijms25158263] [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: 06/28/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Nowadays, GSK3 is accepted as an enzyme strongly involved in the regulation of inflammation by balancing the pro- and anti-inflammatory responses of cells and organisms, thus influencing the initiation, progression, and resolution of inflammatory processes at multiple levels. Disturbances within its broad functional scope, either intrinsically or extrinsically induced, harbor the risk of profound disruptions to the regular course of the immune response, including the formation of severe inflammation-related diseases. Therefore, this review aims at summarizing and contextualizing the current knowledge derived from animal models to further shape our understanding of GSK3α and β and their roles in the inflammatory process and the occurrence of tissue/organ damage. Following a short recapitulation of structure, function, and regulation of GSK3, we will focus on the lessons learned from GSK3α/β knock-out and knock-in/overexpression models, both conventional and conditional, as well as a variety of (predominantly rodent) disease models reflecting defined pathologic conditions with a significant proportion of inflammation and inflammation-related tissue injury. In summary, the literature suggests that GSK3 acts as a crucial switch driving pro-inflammatory and destructive processes and thus contributes significantly to the pathogenesis of inflammation-associated diseases.
Collapse
Affiliation(s)
| | | | | | - René Huber
- Institute of Clinical Chemistry and Laboratory Medicine, Hannover Medical School, 30625 Hannover, Germany; (F.K.); (K.B.); (R.L.)
| |
Collapse
|
18
|
Boengler K, Eickelmann C, Kleinbongard P. Mitochondrial Kinase Signaling for Cardioprotection. Int J Mol Sci 2024; 25:4491. [PMID: 38674076 PMCID: PMC11049936 DOI: 10.3390/ijms25084491] [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: 03/01/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Myocardial ischemia/reperfusion injury is reduced by cardioprotective adaptations such as local or remote ischemic conditioning. The cardioprotective stimuli activate signaling cascades, which converge on mitochondria and maintain the function of the organelles, which is critical for cell survival. The signaling cascades include not only extracellular molecules that activate sarcolemmal receptor-dependent or -independent protein kinases that signal at the plasma membrane or in the cytosol, but also involve kinases, which are located to or within mitochondria, phosphorylate mitochondrial target proteins, and thereby modify, e.g., respiration, the generation of reactive oxygen species, calcium handling, mitochondrial dynamics, mitophagy, or apoptosis. In the present review, we give a personal and opinionated overview of selected protein kinases, localized to/within myocardial mitochondria, and summarize the available data on their role in myocardial ischemia/reperfusion injury and protection from it. We highlight the regulation of mitochondrial function by these mitochondrial protein kinases.
Collapse
Affiliation(s)
- Kerstin Boengler
- Institute of Physiology, Justus-Liebig University, 35392 Giessen, Germany
| | - Chantal Eickelmann
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, 45147 Essen, Germany; (C.E.); (P.K.)
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, 45147 Essen, Germany; (C.E.); (P.K.)
| |
Collapse
|
19
|
Liang J, Yu M, Li Y, Zhao L, Wei Q. Glycogen synthase kinase-3: A potential immunotherapeutic target in tumor microenvironment. Biomed Pharmacother 2024; 173:116377. [PMID: 38442671 DOI: 10.1016/j.biopha.2024.116377] [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: 12/10/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/07/2024] Open
Abstract
Glycogen synthase kinase-3(GSK-3) is a protein kinase that can phosphorylate over a hundred substrates and regulate cell differentiation, proliferation, and death. Researchers have acknowledged the pivotal role of abnormal activation of GSK-3 in the progression of various diseases over the past few decades. Recent studies have mostly concentrated on investigating the function of GSK-3 in the tumor microenvironment, specifically examining the interaction between TAM, NK cells, B cells, and T cells. Furthermore, GSK-3 exhibits a strong association with immunological checkpoints, such as programmed cell death protein 1. Novel GSK-3 inhibitors have potential in tumor immunotherapy, exerting beneficial effects on hematologic diseases and solid tumors. Nevertheless, there is a lack of reviews about the correlation between tumor-associated immune cells and GSK-3. This study intends to analyze the function and mechanism of GSK-3 comprehensively and systematically in the tumor microenvironment, with a special focus on its influence on various immune cells. The objective is to present novel perspectives for GSK-3 immunotherapy.
Collapse
Affiliation(s)
- Jingyi Liang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China; Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning Province 110122, China.
| | - Meng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Yunong Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China; Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning Province 110122, China.
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China; Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning Province 110122, China.
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China; Liaoning Province, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning Province 110122, China.
| |
Collapse
|
20
|
Liang LL, He MF, Zhou PP, Pan SK, Liu DW, Liu ZS. GSK3β: A ray of hope for the treatment of diabetic kidney disease. FASEB J 2024; 38:e23458. [PMID: 38315453 DOI: 10.1096/fj.202302160r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/09/2023] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
Diabetic kidney disease (DKD), a major microvascular complication of diabetes, is characterized by its complex pathogenesis, high risk of chronic renal failure, and lack of effective diagnosis and treatment methods. GSK3β (glycogen synthase kinase 3β), a highly conserved threonine/serine kinase, was found to activate glycogen synthase. As a key molecule of the glucose metabolism pathway, GSK3β participates in a variety of cellular activities and plays a pivotal role in multiple diseases. However, these effects are not only mediated by affecting glucose metabolism. This review elaborates on the role of GSK3β in DKD and its damage mechanism in different intrinsic renal cells. GSK3β is also a biomarker indicating the progression of DKD. Finally, the protective effects of GSK3β inhibitors on DKD are also discussed.
Collapse
Affiliation(s)
- Lu-Lu Liang
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, P.R. China
- Henan Province Research Center For Kidney Disease, Zhengzhou, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, P.R. China
| | - Meng-Fei He
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, P.R. China
- Henan Province Research Center For Kidney Disease, Zhengzhou, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, P.R. China
| | - Pan-Pan Zhou
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, P.R. China
- Henan Province Research Center For Kidney Disease, Zhengzhou, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, P.R. China
| | - Shao-Kang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, P.R. China
- Henan Province Research Center For Kidney Disease, Zhengzhou, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, P.R. China
| | - Dong-Wei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, P.R. China
- Henan Province Research Center For Kidney Disease, Zhengzhou, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, P.R. China
| | - Zhang-Suo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, P.R. China
- Henan Province Research Center For Kidney Disease, Zhengzhou, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, P.R. China
| |
Collapse
|
21
|
Song Y, Kim N, Heo J, Shum D, Heo T, Seo HR. Inhibition of DNMT3B expression in activated hepatic stellate cells overcomes chemoresistance in the tumor microenvironment of hepatocellular carcinoma. Sci Rep 2024; 14:115. [PMID: 38168140 PMCID: PMC10761987 DOI: 10.1038/s41598-023-50680-6] [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: 02/22/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a complex disease associated with a plethora of environmental and genetic/hereditary causative risk factors, more so than other oncological indications. Additionally, patients with HCC exhibit fibrosis, cirrhosis, and liver-related disease. This complicated etiology can affect the disease course and likely contributes to its poor prognosis. In this study, we aimed to improve HCC therapy by evaluating combination treatment using anti-cancer and anti-fibrosis drugs via identification of novel anti-fibrosis drugs. We performed high-throughput screening of 10,000 compounds to identify hepatic fibrosis inhibitors through morphometry analysis of multicellular hepatic spheroid (MCHS) models and identified CHIR-99021 as a candidate anti-fibrotic drug. Treatment with CHIR-99021 induced loss of cell-cell interactions and suppression of extracellular matrix-related protein expression via reprogramming of hepatic stellate cell (HSC) activation in MCHSs. In particular, CHIR-99021 regulated DNMT3B expression only in activated HSCs. Moreover, CHIR-99021 markedly improved the efficacy of sorafenib in HCC- multicellular tumor spheroids in vitro and through induction of apoptosis by decreasing DNMT3B expression in vivo. In summary, these findings suggest that targeting HSC reprogramming by attenuation of DNMT3B expression in the tumor environment might represent a promising therapeutic strategy for liver fibrosis and HCC.
Collapse
Affiliation(s)
- Yeonhwa Song
- Advanced Biomedical Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Namjeong Kim
- Advanced Biomedical Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Jinyeong Heo
- Screening Discovery Platform, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - David Shum
- Screening Discovery Platform, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Taemoo Heo
- Advanced Biomedical Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Haeng Ran Seo
- Advanced Biomedical Research Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
22
|
Yang X, Liang J, Shu Y, Wei L, Wen C, Luo H, Ma L, Qin T, Wang B, Zeng S, Liu Y, Zhou C. Asperosaponin VI facilitates the regeneration of skeletal muscle injury by suppressing GSK-3β-mediated cell apoptosis. J Cell Biochem 2024; 125:115-126. [PMID: 38079224 DOI: 10.1002/jcb.30510] [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: 07/12/2023] [Revised: 10/26/2023] [Accepted: 11/26/2023] [Indexed: 01/16/2024]
Abstract
Asperosaponin VI (ASA VI) is a bioactive triterpenoid saponin extracted from Diptychus roots, of Diptyl, and has previously shown protective functions in rheumatoid arthritis and sepsis. This study investigates the effects and molecular mechanisms of ASA VI on skeletal muscle regeneration in a cardiotoxin (CTX)-induced skeletal muscle injury mouse model. Mice were subjected to CTX-induced injury in the tibialis anterior and C2C12 myotubes were treated with CTX. Muscle fiber histology was analyzed at 7 and 14 days postinjury. Apoptosis and autophagy-related protein expression were evaluated t s by Western blot, and muscle regeneration markers were quantified by quantitative polymerase chain reaction. Docking studies, cell viability assessments, and glycogen synthase kinase-3β (GSK-3β) activation analyses were performed to elucidate the mechanism. ASA VI was observed to improve muscle interstitial fibrosis, remodeling, and performance in CTX-treated mice, thereby increased skeletal muscle size, weight, and locomotion. Furthermore, ASA VI modulated the expression of apoptosis and autophagy-related proteins through GSK-3β inhibition and activated the transcription of regeneration genes. Our results suggest that ASA VI mitigates skeletal muscle injury by modulating apoptosis and autophagy via GSK-3β signaling and promotes regeneration, thus presenting a probable therapeutic agent for skeletal muscle injury.
Collapse
Affiliation(s)
- Xinru Yang
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jian Liang
- Department of Pediatrics, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yue Shu
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Linlin Wei
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Cailing Wen
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Hui Luo
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Liqing Ma
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Tian Qin
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Bin Wang
- Department of Cardiovascular Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Siyu Zeng
- Department of Pharmacy, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ying Liu
- Department of Pharmacology, School of Pharmacy, Macau University of Science and Technology, Taipa, Macao, China
- Department of Pharmacology, School of Pharmacy, Guangzhou Xinhua University, Guangzhou, China
| | - Chun Zhou
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
23
|
Mozaffari MS. Serum Glucocorticoid-Regulated Kinase-1 in Ischemia-Reperfusion Injury: Blessing or Curse. J Pharmacol Exp Ther 2023; 387:277-287. [PMID: 37770199 DOI: 10.1124/jpet.123.001846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 10/03/2023] Open
Abstract
The family of serum-glucocorticoid-regulated kinase (SGK) consists of three paralogs, SGK-1, SGK-2, and SGK-3, with SGK-1 being the better studied. Indeed, recognition of the role of SGK-1 in regulation of cell survival and proliferation has led to introduction of a number of small-molecule inhibitors for some types of cancer. In addition, SGK-1 regulates major physiologic effects, such as renal solute transport, and contributes to the pathogenesis of non-neoplastic conditions involving major organs including the heart and the kidney. These observations raise the prospect for therapeutic modulation of SGK-1 to reduce the burden of such diseases as myocardial infarction and acute kidney injury. Following a brief description of the structure and function of SGK family of proteins, the present review is primarily focused on our current understanding of the role of SGK-1 in pathologies related to ischemia-reperfusion injury involving several organs (e.g., heart, kidney). The essential role of the mitochondrial permeability transition pore in cell death coupled with the pro-survival function of SGK-1 raise the prospect that its therapeutic modulation could beneficially impact conditions associated with ischemia-reperfusion injury. SIGNIFICANCE STATEMENT: Since the discovery of serum glucocorticoid-regulated kinase (SGK)-1, extensive research has unraveled its role in cancer biology and, thus, its therapeutic targeting. Increasingly, it is also becoming clear that SGK-1 is a major determinant of the outcome of ischemia-reperfusion injury to various organs. Thus, evaluation of existing information should help identify gaps in our current knowledge and also determine whether and how its therapeutic modulation could impact the outcome of ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Mahmood S Mozaffari
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
24
|
Tocantins C, Martins JD, Rodrigues ÓM, Grilo LF, Diniz MS, Stevanovic-Silva J, Beleza J, Coxito P, Rizo-Roca D, Santos-Alves E, Rios M, Carvalho L, Moreno AJ, Ascensão A, Magalhães J, Oliveira PJ, Pereira SP. Metabolic mitochondrial alterations prevail in the female rat heart 8 weeks after exercise cessation. Eur J Clin Invest 2023; 53:e14069. [PMID: 37525474 DOI: 10.1111/eci.14069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/15/2023] [Accepted: 06/30/2023] [Indexed: 08/02/2023]
Abstract
BACKGROUND The consumption of high-caloric diets strongly contributes to the development of non-communicable diseases (NCDs), including cardiovascular disease, the leading cause of mortality worldwide. Exercise (along with diet intervention) is one of the primary non-pharmacological approaches to promote a healthier lifestyle and counteract the rampant prevalence of NCDs. The present study evaluated the effects of exercise cessation after a short period training on the cardiac metabolic and mitochondrial function of female rats. METHODS Seven-week-old female Sprague-Dawley rats were fed a control or a high-fat, high-sugar (HFHS) diet and, after 7 weeks, the animals were kept on a sedentary lifestyle or submitted to endurance exercise for 3 weeks (6 days per week, 20-60 min/day). The cardiac samples were analysed 8 weeks after exercise cessation. RESULTS The consumption of the HFHS diet triggered impaired glucose tolerance, whereas the HFHS diet and physical exercise resulted in different responses in plasma adiponectin and leptin levels. Cardiac mitochondrial respiration efficiency was decreased by the HFHS diet consumption, which led to reduced ATP and increased NAD(P)H mitochondrial levels, which remained prevented by exercise 8 weeks after cessation. Exercise training-induced cardiac adaptations in redox balance, namely increased relative expression of Nrf2 and downstream antioxidant enzymes persist after an eight-week exercise cessation period. CONCLUSIONS Endurance exercise modulated cardiac redox balance and mitochondrial efficiency in female rats fed a HFHS diet. These findings suggest that exercise may elicit cardiac adaptations crucial for its role as a non-pharmacological intervention for individuals at risk of developing NCDs.
Collapse
Affiliation(s)
- Carolina Tocantins
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - João D Martins
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Óscar M Rodrigues
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Luís F Grilo
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Mariana S Diniz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Jelena Stevanovic-Silva
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Jorge Beleza
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - David Rizo-Roca
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Estela Santos-Alves
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Manoel Rios
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Lina Carvalho
- Institute of Anatomical and Molecular Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - António J Moreno
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, School of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| | - Paulo J Oliveira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Susana P Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
| |
Collapse
|
25
|
Shin HK, Seo KJ, Lee JY, Jeon SR, Yune TY. GSK-3β and β-Catenin Signaling Pathway is Involved in Myofibroblast Transition of Ligamentum Flavum in Lumbar Spinal Stenosis Patients. Spine (Phila Pa 1976) 2023; 48:1472-1479. [PMID: 37417723 DOI: 10.1097/brs.0000000000004770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/25/2023] [Indexed: 07/08/2023]
Abstract
STUDY DESIGN Histologic analysis of the ligamentum flavum (LF) in the lumbar spine. OBJECTIVE The objective of this study is to investigate the levels of glycogen synthase kinase-3β (GSK-3β) and β-catenin in the LF tissue of patients with lumbar spinal stenosis (LSS). SUMMARY OF BACKGROUND DATA The hypertrophy of the LF is the primary cause of the progression of LSS. Recently, Wnt signaling has been proposed as one of the molecular processes contributing to LF hypertrophy. GSK-3β and β-catenin are recognized to play a crucial part in the control of this signaling pathway. MATERIALS AND METHODS From May 2020 to July 2022, LF from 51 LSS patients (LSS group) and 18 lumbar disc herniation patients (control group) were prospectively collected during surgery. Histologic analysis was investigated to confirm the progression of LF fibrosis. The levels of α-smooth muscle actin, phosphorylation of GSK-3β (p-GSK-3β; inactive form), and β-catenin were analyzed in LF with Western blot analysis to reveal the GSK-3β/β-catenin signaling pathway. Continuous variables are expressed as mean±SD and compared using the student t test. Categorical variables are compared using the χ 2 test or Fisher exact test, as appropriate. To determine the association between p-GSK-3β and LF thickness, the Pearson correlation coefficient was calculated based on the results of Western blot analysis. RESULTS The LSS group was older and had thicker LF than the controls. The LSS group showed increased collagen fiber and cellularity than the controls. The levels of α-smooth muscle actin, p-GSK-3β, and β-catenin in the LF of the LSS group were significantly higher than that of the control group. There was a strong positive correlation between p-GSK-3β (Ser9) level and LF thickness in LSS patients ( r =0.69, P =0.01). CONCLUSION This research proposes a molecular mechanism for the pathogenesis of LF hypertrophy in LSS. Specifically, GSK-3β/β-catenin signaling appears to be related to LF hypertrophy in LSS and a positive correlation exists between p-GSK-3β level and LF thickness. LEVEL OF EVIDENCE Level 3.
Collapse
Affiliation(s)
- Hong Kyung Shin
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung Jin Seo
- Department Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Jee Youn Lee
- Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul, Republic of Korea
| | - Sang Ryong Jeon
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Tae Young Yune
- Department Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul, Republic of Korea
- Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Trionfetti F, Marchant V, González-Mateo GT, Kawka E, Márquez-Expósito L, Ortiz A, López-Cabrera M, Ruiz-Ortega M, Strippoli R. Novel Aspects of the Immune Response Involved in the Peritoneal Damage in Chronic Kidney Disease Patients under Dialysis. Int J Mol Sci 2023; 24:5763. [PMID: 36982834 PMCID: PMC10059714 DOI: 10.3390/ijms24065763] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Chronic kidney disease (CKD) incidence is growing worldwide, with a significant percentage of CKD patients reaching end-stage renal disease (ESRD) and requiring kidney replacement therapies (KRT). Peritoneal dialysis (PD) is a convenient KRT presenting benefices as home therapy. In PD patients, the peritoneum is chronically exposed to PD fluids containing supraphysiologic concentrations of glucose or other osmotic agents, leading to the activation of cellular and molecular processes of damage, including inflammation and fibrosis. Importantly, peritonitis episodes enhance peritoneum inflammation status and accelerate peritoneal injury. Here, we review the role of immune cells in the damage of the peritoneal membrane (PM) by repeated exposure to PD fluids during KRT as well as by bacterial or viral infections. We also discuss the anti-inflammatory properties of current clinical treatments of CKD patients in KRT and their potential effect on preserving PM integrity. Finally, given the current importance of coronavirus disease 2019 (COVID-19) disease, we also analyze here the implications of this disease in CKD and KRT.
Collapse
Affiliation(s)
- Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
| | - Vanessa Marchant
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
- REDINREN/RICORS2040, 28029 Madrid, Spain
| | - Guadalupe T. González-Mateo
- Cell-Cell Communication & Inflammation Unit, Centre for Molecular Biology “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain
- Premium Research, S.L., 19005 Guadalajara, Spain
| | - Edyta Kawka
- Department of Pathophysiology, Poznan University of Medical Sciences, 10 Fredry St., 61-701 Poznan, Poland
| | - Laura Márquez-Expósito
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
- REDINREN/RICORS2040, 28029 Madrid, Spain
| | - Alberto Ortiz
- IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Manuel López-Cabrera
- Cell-Cell Communication & Inflammation Unit, Centre for Molecular Biology “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
- REDINREN/RICORS2040, 28029 Madrid, Spain
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
| |
Collapse
|
27
|
Guo J, Zheng W, Liu Y, Zhou M, Shi Y, Lei M, Zhang C, Liu Z. Long non-coding RNA DLX6-AS1 is the key mediator of glomerular podocyte injury and albuminuria in diabetic nephropathy by targeting the miR-346/GSK-3β signaling pathway. Cell Death Dis 2023; 14:172. [PMID: 36854759 PMCID: PMC9975222 DOI: 10.1038/s41419-023-05695-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 03/02/2023]
Abstract
Progressive albuminuria is the primary clinical symptom of diabetic nephropathy (DN), leading to a gradual decline in kidney function. DLX6-AS1 was the first reported long non-coding RNA (lncRNA) to participate in organogenesis and play crucial roles in the brain or neural cell development. Herein, we investigated the DLX6-AS1 (Dlx6-os1 in mice) role in DN pathogenesis. We found that DLX6-AS1 expression in DN patients correlated with the extent of albuminuria. Dlx6-os1 overexpression induced cellular damage and inflammatory responses in cultured podocytes through miR-346-mediated regulation of the GSK-3β pathway. In various established diabetic and newly developed knockout mouse models, Dlx6-os1 knockdown/knockout significantly reduced podocyte injury and albuminuria. The Dlx6-os1 effects were remarkably modulated by miR-346 mimics or mutants and significantly diminished in podocyte-specific GSK-3β-knockout mice. Thus, DLX6-AS1 (Dlx6-os1) promotes DN development by accelerating podocyte injury and inflammation through the upregulation of the GSK-3β pathway, providing a novel molecular target for DN therapy.
Collapse
Affiliation(s)
- Jia Guo
- Nephrology Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China.
| | - Wen Zheng
- Nephrology Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Yong Liu
- Nephrology Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Mengwen Zhou
- Nephrology Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Yan Shi
- Nephrology Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Min Lei
- Nephrology Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Chaojie Zhang
- Nephrology Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Zhangsuo Liu
- Nephrology Research Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China.
| |
Collapse
|
28
|
Guo Q, Wu D, Jia D, Zhang X, Wu A, Lou L, Zhao M, Zhao M, Gao Y, Wang M, Liu M, Chen M, Zhang D. Bioinformatics prediction and experimental verification of a novel microRNA for myocardial fibrosis after myocardial infarction in rats. PeerJ 2023; 11:e14851. [PMID: 36788811 PMCID: PMC9922498 DOI: 10.7717/peerj.14851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/13/2023] [Indexed: 02/11/2023] Open
Abstract
Background MicroRNAs (miRNAs) are endogenous noncoding single-stranded small RNAs. Numerous studies have shown that miRNAs have pivotal roles in the occurrence and development of myocardial fibrosis (MF). However, miRNA expression profile in rats with MF after myocardial infarction (MI) is not well understood. The present study aimed to find the potential miRNA for MF post MI. Methods SPF male Sprague-Dawley (SD) rat models of acute myocardial infarction (AMI) were established by ligating the anterior descending branch of the left coronary artery, while sham-operated rats were only threaded without ligation as a control group. Hematoxylin-eosin and Masson trichrome staining were used to detect myocardial histopathological changes for model evaluation. The differentially expressed miRNAs were detected by using the Agilent Rat miRNA gene chip in the myocardial tissue of the infarct marginal zone. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed by DAVID. The expression of miR-199a-5p was verified by real-time fluorescence quantitative PCR (qRT-PCR). Transfected miR-199a-5p mimics into cardiac fibroblasts (CFs) to construct cell models of miR-199a-5p overexpression. Dual-luciferase reporter assay was employed to validate the target gene of miR-199a-5p. The protein expression of the target gene in CFs transfected with miR-199a-5p mimics were detected by Western blot. Results Myocardial fibrosis was exacerbated in the model group compared with the control group. Thirteen differentially expressed miRNAs between the two groups were screened and their expression levels in the model group were all higher than those in the control group. The expression of miR-199a-5p was significantly increased in the model group in qRT-PCR, which was consistent with the results of the gene chip. KEGG enrichment analysis showed that the target genes of miR-199a-5p were enriched in the insulin signaling pathway. Furthermore, dual-luciferase reporter assay indicated that miR-199a-5p could negatively regulate the expression of GSK-3β. After transfection, the expression of miR-199a-5p was increased in the miR-199a-5p mimics group. The protein expression of GSK-3β was decreased in CFs transfected with miR-199a-5p mimics. Conclusion Our study identified miR-199a-5p could promote the progression of myocardial fibrosis after myocardial infarction by targeting GSK-3β, which provides novel targets for diagnosis and treatment of MF.
Collapse
Affiliation(s)
- Qianqian Guo
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dandan Wu
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Dongdong Jia
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyue Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Aiming Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lixia Lou
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mengzhu Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yijie Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Manman Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Menghua Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Meng Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Dongmei Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
29
|
Huang Y, Zhang Y, Wu Y, Xiang Q, Yu R. An Integrative Pharmacology-Based Strategy to Uncover the Mechanism of Zuogui Jiangtang Shuxin Formula in Diabetic Cardiomyopathy. Drug Des Devel Ther 2023; 17:237-260. [PMID: 36726736 PMCID: PMC9885885 DOI: 10.2147/dddt.s390883] [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: 09/30/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023] Open
Abstract
Purpose This study aimed to explore the mechanism of Zuogui Jiangtang Shuxin formula (ZGJTSXF) in the treatment of diabetic cardiomyopathy (DCM) by an integrative strategy combining serum pharmacochemistry, network pharmacology analysis, and experimental validation. Methods An Ultra high performance liquid chromatography-high resolution mass spectrometry (UPLC-Q-Exactive-Orbitrap-MS) method was constructed to identify compounds in rat serum after oral administration of ZGJTSXF. A component-target network between the targets of ZGJTSXF ingredients and DCM was established using Cytoscape. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed to deduce ZGJTSXF-associated targets and pathways. The DCM model mice were treated with ZGJTSXF, and the predicted important signaling pathways were verified using quantitative PCR and Western blot. Results We identified 78 compounds in serum of medicated rats, which mainly included flavonoids, small peptides, nucleosides, organic acids, phenylpropanoids, alkaloids, phenanthrenequinones, iridoids, phenols, and saponins. Network pharmacology analysis revealed that ZGJTSXF may regulate targets including ALB, TNF, AKT1, GAPDH, VEGFA, EGFR, SRC, CASP3, MAPK3, JUN, and PI3K/AKT signaling pathway in the treatment of DCM. ZGJTSXF administration improved blood sugar levels, heart function, and cardiac morphological changes in DCM mice. Notably, ZGJTSXF inhibited cardiomyocytes apoptosis, which was associated with restored PI3K/Akt signaling and upregulated Bcl-2 and Bcl-xL proteins expression. Conclusion Our preliminary results proposed the material basis and possible mechanisms of ZGJTSXF in treating DCM, which is related to the activation of the PI3K/AKT signaling pathway and apoptosis inhibition. These findings shed new light in developing ZGJTSXF-based therapeutics in treating DCM.
Collapse
Affiliation(s)
- Yalan Huang
- Graduate School, Hunan University of Traditional Chinese Medicine, Changsha, 410208, People’s Republic of China,The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, 410021, People’s Republic of China
| | - Yanling Zhang
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, 410208, People’s Republic of China,General Hospital of Ningxia Medical University, Ningxia, 750003, People’s Republic of China
| | - Yongjun Wu
- College of Pharmacy, Hunan University of Traditional Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Qin Xiang
- Science and Technology Department, Hunan University of Traditional Chinese Medicine, Changsha, 410208, People’s Republic of China,Qin Xiang, Science and Technology Department, Hunan University of Traditional Chinese Medicine, Changsha, 410208, People’s Republic of China, Email
| | - Rong Yu
- Graduate School, Hunan University of Traditional Chinese Medicine, Changsha, 410208, People’s Republic of China,Correspondence: Rong Yu, Graduate School, Hunan University of Traditional Chinese Medicine, Changsha, 410208, People’s Republic of China, Email
| |
Collapse
|
30
|
Wu Y, Bai Y, Feng Y, Zhang Q, Diao Z, Liu W. Renalase Prevents Renal Fibrosis by Inhibiting Endoplasmic Reticulum Stress and Down-Regulating GSK-3β/Snail Signaling. Int J Med Sci 2023; 20:669-681. [PMID: 37082730 PMCID: PMC10110476 DOI: 10.7150/ijms.82192] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/24/2023] [Indexed: 04/22/2023] Open
Abstract
Background: Treating renal fibrosis is crucial to delaying chronic kidney disease. The glycogen synthase kinase-3β (GSK-3β)/Snail pathway regulates renal fibrosis and Renalase can ameliorate renal interstitial fibrosis. However, it is not clear whether GSK-3β/Snail signaling affects Renalase action. Here, we explored the role and mechanism of GSK-3β/Snail in the anti-fibrosis action of Renalase. Materials and methods: We used mice with complete unilateral ureteral obstruction (UUO) and human proximal renal tubular epithelial (HK-2) cells with transforming growth factor-β1 (TGF-β1)-induced fibrosis to explore the role and regulatory mechanism of the GSK-3β/Snail pathway in the amelioration of renal fibrosis by Renalase. Results: In UUO mice and TGF-β1-induced fibrotic HK-2 cells, the expression of p-GSK-3β-Tyr216/p-GSK-3β-Ser9, GSK-3β and Snail was significantly increased, and endoplasmic reticulum (ER) stress was activated. After Renalase supplementation, fibrosis was alleviated, ER stress was inhibited and p-GSK-3β-Tyr216/p-GSK-3β-Ser9, GSK-3β and Snail were significantly down-regulated. The amelioration of renal fibrosis by Renalase and its inhibitory effect on GSK-3β/Snail were reversed by an ER stress agonist. Furthermore, when an adeno-associated virus or plasmid was used to overexpress GSK-3β, the effect of Renalase on delaying renal fibrosis was counteracted, although ER stress markers did not change. Conclusion: Renalase prevents renal fibrosis by down-regulating GSK-3β/Snail signaling through inhibition of ER stress. Exogenous Renalase may be an effective method of slowing or stopping chronic kidney disease progression.
Collapse
Affiliation(s)
- Yiru Wu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing 100050, P. R. China
| | - Yu Bai
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing 100050, P. R. China
| | - Yiduo Feng
- Department of Nephrology, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring Road West, Fengtai District, Beijing,100070, P. R. China
| | - Qidong Zhang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing 100050, P. R. China
| | - Zongli Diao
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing 100050, P. R. China
| | - Wenhu Liu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xi Cheng District, Beijing 100050, P. R. China
- ✉ Corresponding author: Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P. R. China. Tel.: +86-10-63138579; Fax: +86-10-63139144; E-mail:
| |
Collapse
|
31
|
Du C, Dong J, Wang Q, Xu C, Feng S, Feng R, Lv X, Li J, Zhang L, Huang C, Ma T. Hastatoside attenuatescarbon tetrachloride-induced liver fibrosis by targeting glycogen synthase kinase-3β. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154585. [PMID: 36610117 DOI: 10.1016/j.phymed.2022.154585] [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: 05/29/2022] [Revised: 11/11/2022] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Hastatoside is an iridoid glycoside extracted from the herb, Verbena officinalis, that exerts various pharmacological effects, including anti-inflammatory, sleep-promoting, and analgesic effects. However, only a few studies have reported the efficacy of hastatoside in liver fibrosis. Liver fibrosis is a pathophysiological process, and its persistence can seriously affect the quality of life and well-being of the patients. HYPOTHESIS/PURPOSE This study aimed to investigate the role of hastatoside on liver fibrosis and its possible underlying mechanisms. METHODS C57BL/6 J mice with carbon tetrachloride (CCl4)-induced hepatic fibrosis were used as the in vivo models. Histological features of the liver were observed using Masson's trichrome and hematoxylin-eosin staining. Alanine aminotransferase and aspartate aminotransferase levels and the hepatic fibrosis indices (type 3 procollagen, laminin, and hyaluronic acid) were measured using corresponding assay kits. LX-2 human hepatic stellate cells (HSCs) stimulated with the transforming growth factor β1 were used as the vitro models. Transfection of the glycogen synthase kinase (GSK)-3β small interfering RNA (siRNA) and β-catenin plasmids was also performed in vitro. Protein levels of GSK-3β, phospho-GSK-3β (Ser 9), α-smooth muscle actin, collagen type I alpha 1, c-Myc, cyclin D1, and β-catenin were determined via western blotting. Moreover, the p-GSK-3β:GSK-3β ratio was calculated to determine the GSK-3β activity. RESULTS Hastatoside prevented CCl4-induced liver injury and histological damage. It inhibited the upregulation of α-SMA and Col1α1 levels in a CCl4-induced mouse hepatic fibrosis model. In vitro, hastatoside inhibited the proliferation and activation of HSCs by decreasing the expression levels of cyclin D1 and c-Myc and the proportion of LX-2 cells activated in the G0/G1 phase. Molecular docking results showed that hastatoside bound to GSK-3β. Hastatoside significantly increased the GSK-3β activity and inhibited the downstream effector expression of β-catenin. CONCLUSION These findings suggest that hastatoside can bind to GSK-3β and promote its activity, while inhibiting the GSK-3β downstream effector expression of β-catenin, thereby inhibiting the activation and proliferation of HSCs, which further prevents the development of liver fibrosis. These results provide innovative insights into the underlying liver fibrosis. Moreover, hastatoside is a potential anti-fibrosis monomer that can potentially be used for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Changlin Du
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, China
| | - Jiahui Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, China
| | - Qi Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, China
| | - Chuanting Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, China
| | - Shiqi Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, China
| | - Rui Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, China
| | - Xiongwen Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Lei Zhang
- Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, China.
| | - Taotao Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China; Center of Traditional Chinese Medicine Formula Granule, Anhui Medical University, China.
| |
Collapse
|
32
|
Guan Z, Wang Y, Xu H, Wang Y, Wu D, Zhang Z, Liu Z, Shang N, Zhang D, Sun J, He X, Li Y, Zhu L, Liu Z, Zhang M, Xu Z, Song Z, Dai G. Isoandrographolide from Andrographis paniculata ameliorates tubulointerstitial fibrosis in ureteral obstruction-induced mice, associated with negatively regulating AKT/GSK-3β/β-cat signaling pathway. Int Immunopharmacol 2022; 112:109201. [PMID: 36067652 DOI: 10.1016/j.intimp.2022.109201] [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: 05/21/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/05/2022]
Abstract
Tubulointerstitial fibrosis (TIF) is a prominent pathological manifestation for the progression of almost all chronic kidney diseases (CKDs) to end-stage renal failure. However, there exist few efficient therapies to cure TIF. Our recent results showed that (8R, 12S)-isoandrographolide (ISA), a diterpenoid lactone ingredient of traditional Chinese herbal Andrographis paniculata (Burm.f.) Nees, exhibited anti-pulmonary fibrosis in silica-induced mice. Herein, we investigated the therapeutic effect of ISA on TIF, using mice subjected to unilateral ureteral obstruction (UUO) and human kidney proximal tubular epithelial (HK-2) cells treated with transforming growth factor-β1 (TGF-β1) or tumor necrosis factor-α (TNF-α). The pathological changes and collagen deposition results displayed that ISA administration significantly attenuated inflammatory response, ameliorated TIF, and protected the kidney injury. Interestingly, ISA revealed much lower cytotoxicity on HK-2 cells, but exhibited stronger inhibitory effect on tubular epithelial mesenchymal transformation (EMT) and inflammation, as compared to andrographolide (AD), the major ingredient of A. paniculata extract that has been reported to ameliorate TIF in diabetic nephropathy mice. It was further clarified that the amelioration of TIF by ISA was associated with suppressing the aberrant activation of AKT/GSK-3β/β-catenin pathway through network pharmacology analysis and experimental validation. Taken together, these findings indicate that ISA is a promising lead compound for development of anti-TIF, and even broad-spectrum anti-fibrotic drugs.
Collapse
Affiliation(s)
- Zhenzhen Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Yaming Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Haiwei Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Yake Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Di Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Zhizi Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Zihan Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Ning Shang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Di Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Jingyang Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Xugang He
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Yingxue Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Lina Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Zhentao Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Mingliang Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Zhihao Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China
| | - Zhe Song
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China.
| | - Guifu Dai
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, People's Republic of China.
| |
Collapse
|
33
|
Cai S, Hu T, Venkatesan M, Allam M, Schneider F, Ramalingam SS, Sun SY, Coskun AF. Multiplexed protein profiling reveals spatial subcellular signaling networks. iScience 2022; 25:104980. [PMID: 36093051 PMCID: PMC9460555 DOI: 10.1016/j.isci.2022.104980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 05/25/2022] [Accepted: 08/16/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Shuangyi Cai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Thomas Hu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Mythreye Venkatesan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Mayar Allam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Frank Schneider
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, GA 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Suresh S. Ramalingam
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shi-Yong Sun
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ahmet F. Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Corresponding author
| |
Collapse
|
34
|
Cai J, Wang T, Zhou Y, Tang C, Liu Y, Dong Z. Phosphorylation by GSK-3β increases the stability of SIRT6 to alleviate TGF-β-induced fibrotic response in renal tubular cells. Life Sci 2022; 308:120914. [PMID: 36057401 DOI: 10.1016/j.lfs.2022.120914] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/16/2022] [Accepted: 08/24/2022] [Indexed: 10/31/2022]
Abstract
AIMS The deacetylase Sirtuin 6 (SIRT6) is up-regulated during fibrogenesis in renal tubular cells and post-ischemia/reperfusion kidneys. Hence, our aim was to investigate the mechanism of SIRT6 up-regulation upon profibrotic stress. MAIN METHODS Immunohistochemical staining was used to detect the expression of UBC9 in the kidney section. The interaction of GSK-3β and SIRT6, and phosphorylation level of SIRT6 were detected by the immunoprecipitation assay. The wild-type and phosphorylated site mutant plasmids of SIRT6 were constructed and stably transfected to BUMPT cells to evaluate the phosphorylation function of SIRT6 by immunoblotting assay. KEY FINDINGS The phosphorylation of SIRT6 is significantly increased during TGF-β treatment in mouse renal tubular cells. GSK-3β can physically interact with SIRT6 in renal tubular cells, and this interaction is enhanced by TGF-β treatment. Moreover, GSK-3β is the phosphorylation kinase for SIRT6, and phosphorylates SIRT6 at Serine 326 residue to prevent its ubiquitination-mediated proteasomal degradation. Non-phosphorylatable mutant, S326A, of SIRT6, restores β-catenin activation and fibrotic changes in renal tubular cells. SIGNIFICANCE The present study demonstrates that a new mechanism for GSK-3β-mediated anti-fibrotic function in renal fibrosis through phosphorylation of SIRT6 to prevent its proteasomal degradation.
Collapse
Affiliation(s)
- Juan Cai
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Tianshi Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunqian Zhou
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China.
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
35
|
Kim Y, Nam Y, Rim YA, Ju JH. Anti-fibrotic effect of a selective estrogen receptor modulator in systemic sclerosis. Stem Cell Res Ther 2022; 13:303. [PMID: 35841004 PMCID: PMC9284699 DOI: 10.1186/s13287-022-02987-w] [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: 03/14/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022] Open
Abstract
Background The rarity of systemic sclerosis (SSc) has hampered the development of therapies for this intractable autoimmune disease. Induced pluripotent stem cell (iPSC) can be differentiated into the key disease-affected cells in vitro. The generation of patient-derived iPSCs has opened up possibilities for rare disease modeling. Since these cells can recapitulate the disease phenotypes of the cell in question, they are useful high-throughput platforms for screening for drugs that can reverse these abnormal phenotypes. Methods SSc iPSC was generated from PBMC by Sendai virus. Human iPSC lines from SSc patients were differentiated into dermal fibroblasts and keratinocytes. The iPSC-derived differentiated cells from the SSc patients were used on high-throughput platforms to screen for FDA-approved drugs that could be effective treatments for SSc. Results Skin organoids were generated from these cells exhibited fibrosis that resembled SSc skin. Screening of the 770-FDA-approved drug library showed that the anti-osteoporotic drug raloxifene reduced SSc iPSC-derived fibroblast proliferation and extracellular matrix production and skin fibrosis in organoids and bleomycin-induced SSc-model mice. Conclusions This study reveals that a disease model of systemic sclerosis generated using iPSCs-derived skin organoid is a novel tool for in vitro and in vivo dermatologic research. Since raloxifene and bazedoxifene are well-tolerated anti-osteoporotic drugs, our findings suggest that selective estrogen receptor modulator (SERM)-class drugs could treat SSc fibrosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02987-w.
Collapse
Affiliation(s)
- Yena Kim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,YiPSCELL Inc., 47-3, Banpo-dearo 39-gil, Seocho-gu, Seoul, Republic of Korea
| | - Yoojun Nam
- YiPSCELL Inc., 47-3, Banpo-dearo 39-gil, Seocho-gu, Seoul, Republic of Korea
| | - Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea. .,YiPSCELL Inc., 47-3, Banpo-dearo 39-gil, Seocho-gu, Seoul, Republic of Korea. .,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-040, Republic of Korea.
| |
Collapse
|
36
|
Vergoten G, Bailly C. Molecular docking study of GSK-3β interaction with nomilin, kihadanin B, and related limonoids and triterpenes with a furyl-δ-lactone core. J Biochem Mol Toxicol 2022; 36:e23130. [PMID: 35686814 DOI: 10.1002/jbt.23130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 04/01/2022] [Accepted: 05/30/2022] [Indexed: 11/06/2022]
Abstract
Glycogen synthase kinase-3β (GSK-3β) is a target enzyme considered for the treatment of multiple human diseases, from neurodegenerative pathologies to viral infections and cancers. Numerous inhibitors of GSK-3β have been discovered but thus far only a few have reached clinical trials and only one drug, tideglusib (1), has been registered. Natural products targeting GSK-3β have been identified, including the two anticancer limonoids obacunone (5) and gedunin (4), both presenting a furyl-δ-lactone core. To help identifying novel GSK-3β ligands, we have performed a molecular docking study with 15 complementary natural products bearing a furyl-δ-lactone unit (such as limonin (6) and kihadanins A (8) and B (9)) or a closely related structure (such as cedrelone (10) and nimbolide (11)). The formation of GSK-3β-binding complexes for those natural products was compared to reference GSK-3β ATP-competitive inhibitors LY2090314 (3) and AR-A014418 (2). Our in silico analysis led to the identification of two new GSK-3β-binding natural products: kihadanin B (9) and nomilin (7). The latter surpassed the reference compounds in terms of calculated empirical energy of interaction (ΔE). Nomilin (7) can possibly bind to the active site of GSK-3β, notably via the furyl-δ-lactone core and its 1-acetyl group, implicated in the protein interaction. Compound structure-binding relationships are discussed. The study should help the discovery of novel natural products targeting GSK-3β.
Collapse
Affiliation(s)
- Gérard Vergoten
- Inserm, INFINITE - U1286, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), Faculté de Pharmacie, University of Lille, Lille, France
| | - Christian Bailly
- OncoWitan, Scientific Consulting Office, Lille, Wasquehal, France
| |
Collapse
|
37
|
Liu Y, Xu X, Lei W, Hou Y, Zhang Y, Tang R, Yang Z, Tian Y, Zhu Y, Wang C, Deng C, Zhang S, Yang Y. The NLRP3 inflammasome in fibrosis and aging: The known unknowns. Ageing Res Rev 2022; 79:101638. [PMID: 35525426 DOI: 10.1016/j.arr.2022.101638] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/27/2022] [Accepted: 05/01/2022] [Indexed: 11/28/2022]
Abstract
Aging-related diseases such as cancer, cardiovascular diseases, diabetes, and neurodegenerative diseases are often accompanied by fibrosis. The NLRP3 inflammasome triggers the inflammatory response and subsequently promotes fibrosis through pathogen-associated molecular patterns (PAMPs). In this review, we first introduce the general background and specific mechanism of NLRP3 in fibrosis. Second, we investigate the role of NLRP3 in fibrosis in different organs/tissues. Third, we discuss the relationship between NLRP3 and fibrosis during aging. In summary, this review describes the latest progress on the roles of NLRP3 in fibrosis and aging and reveals the possibility of NLRP3 as an antifibrotic and anti-aging treatment target.
Collapse
Affiliation(s)
- Yanqing Liu
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Xuezeng Xu
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Wangrui Lei
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yuxuan Hou
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yan Zhang
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Ran Tang
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Zhi Yang
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Ye Tian
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yanli Zhu
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Changyu Wang
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shaofei Zhang
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Life Sciences and Medicine, Northwest University, Xi'an, China.
| | - Yang Yang
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Life Sciences and Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
38
|
Liu Z, Zhou S, Zhang Y, Zhao M. Rat bone marrow mesenchymal stem cells (BMSCs) inhibit liver fibrosis by activating GSK3β and inhibiting the Wnt3a/β-catenin pathway. Infect Agent Cancer 2022; 17:17. [PMID: 35440002 PMCID: PMC9017036 DOI: 10.1186/s13027-022-00432-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/11/2022] [Indexed: 11/20/2022] Open
Abstract
Background Bone marrow mesenchymal stem cells (BMSCs) can effectively alleviate liver fibrosis, which is a pathological injury caused by various chronic liver diseases. This study aimed to investigate the antifibrotic effects of BMSCs and elucidate the underlying mechanism by which BMSCs affect liver fibrosis in vitro and in vivo. Methods After the rat liver fibrosis model was induced by continuous injection of carbon tetrachloride (CCl4), BMSCs were administered for 4 weeks, and histopathological analysis and liver function tests were performed. T6 hepatic stellate cells (HSC-T6 cells) were stimulated by TGF-β1, and the activation and proliferation of cells were analyzed by CCK-8 assays, flow cytometry, real-time PCR, western blotting and enzyme-linked immunosorbent assay (ELISA). Results Our data demonstrated that BMSCs effectively reduced the accumulation of collagen, enhanced liver functionality and ameliorated liver fibrosis in vivo. BMSCs increased the sub-G1 population in HSC-T6 cells. In addition, coculture with BMSCs reduced the expression of α-SMA, collagen I, cyclin-D1, and c-Myc in HSC-T6 cells and activated the phosphorylation of GSK3β. The GSK3β inhibitor SB216763 reversed the effect of BMSCs. The Wnt/β-catenin signalling pathway was involved in BMSC-mediated inhibition of HSC-T6 cell activation. Conclusions Our data suggested that BMSCs exerted antifibrotic effects by activating the expression of GSK3β and inhibiting the Wnt3a/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Zhaoguo Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China.,Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, China
| | - Song Zhou
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ya Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ming Zhao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China. .,Department of Organ Transplantation, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, Guangdong Province, China.
| |
Collapse
|
39
|
Li Y, Shen Z, Jiang X, Wang Y, Yang Z, Mao Y, Wu Z, Li G, Chen H. Mouse mesenchymal stem cell-derived exosomal miR-466f-3p reverses EMT process through inhibiting AKT/GSK3β pathway via c-MET in radiation-induced lung injury. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:128. [PMID: 35392967 PMCID: PMC8988379 DOI: 10.1186/s13046-022-02351-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/29/2022] [Indexed: 01/06/2023]
Abstract
Background Radiation-induced lung fibrosis (RILF) is a common complication of thoracic radiotherapy. Alveolar epithelial cells play a crucial role in lung fibrosis via epithelial-mesenchymal transition (EMT). Exosomes derived from mesenchymal stem cells own the beneficial properties to repair and regeneration of damaged tissues, however the underlying mechanisms remain poorly understood. Methods Mouse mesenchymal stem cells-derived exosomes (mMSCs-Exo) were isolated by differential centrifugation, and their protective effects were assessed in vivo and in vitro, respectively. EMT-associated proteins were measured via western blot assay and/or immunofluorescence staining. The miRNA expression was measured by microarray assay and qPCR. Furthermore, bioinformatics prediction with KEGG analysis, luciferase assay, and rescue experiments were performed to explore the molecular mechanism underlying miR-466f-3p. Results mMSCs-Exos were efficiently isolated ranging from 90-150 nm with high expression of exosomal markers (CD63, TSG101, and CD9). mMSCs-Exos administration efficiently relieved radiation-induced lung injury with less collagen deposition and lower levels of IL-1β and IL-6. Meanwhile, in vitro results showed mMSCs-Exos treatment obviously reversed EMT process induced by radiation. Among enriched miRNA cargo in exosomes, miR-466f-3p was primarily responsible for the protective effects via inhibition of AKT/GSK3β pathway. Our mechanistic study further demonstrated that c-MET was the direct target of miR-466f-3p, whose restoration partially abrogated mMSCs-Exo-mediated inhibition in both EMT process and AKT/GSK3β signaling activity induced by radiation. Conclusions Our findings indicated that exosomal miR-466f-3p derived from mMSCs may possess anti-fibrotic properties and prevent radiation-induced EMT through inhibition of AKT/GSK3β via c-MET, providing a promising therapeutic modality for radiation-induced lung fibrosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02351-z.
Collapse
Affiliation(s)
- Yi Li
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, 212 Daguan Road, Kunming, 650032, China.
| | - Zhufu Shen
- Department of Geriatrics, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xiao Jiang
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, 212 Daguan Road, Kunming, 650032, China
| | - Yuanyuan Wang
- Department of Pathology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, 650032, China
| | - Zuozhang Yang
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Yuchi Mao
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, 212 Daguan Road, Kunming, 650032, China
| | - Zhixian Wu
- Department of Hepatobiliary Disease, 900th Hospital of Joint Logistics Support Force, Fuzhou, 354200, China
| | - Gaofeng Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China.
| | - Hong Chen
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, 212 Daguan Road, Kunming, 650032, China.
| |
Collapse
|
40
|
Hu X, Zou M, Ni L, Zhang M, Zheng W, Liu B, Cheng Z. Dec1 Deficiency Ameliorates Pulmonary Fibrosis Through the PI3K/AKT/GSK-3β/β-Catenin Integrated Signaling Pathway. Front Pharmacol 2022; 13:829673. [PMID: 35355710 PMCID: PMC8959854 DOI: 10.3389/fphar.2022.829673] [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: 12/06/2021] [Accepted: 02/23/2022] [Indexed: 12/14/2022] Open
Abstract
Tissue remodeling/fibrosis is a main feature of idiopathic pulmonary fibrosis (IPF), which results in the replacement of normal lung parenchyma with a collagen-rich extracellular matrix produced by fibroblasts and myofibroblasts. Epithelial-mesenchymal transition (EMT) in type 2 lung epithelial cells is a key process in IPF, which leads to fibroblasts and myofibroblasts accumulation and excessive collagen deposition. DEC1, a structurally distinct class of basic helix-loop-helix proteins, is associated with EMT in cancer. However, the functional role of DEC1 in pulmonary fibrosis (PF) remains elusive. Herein, we aimed to explore DEC1 expression in IPF and bleomycin (BLM)-induced PF in mice and the mechanisms underlying the fibrogenic effect of DEC1 in PF in vivo and in vitro by Dec1-knockout (Dec1 -/-) mice, knockdown and overexpression of DEC1 in alveolar epithelial cells (A549 cells). We found that the expression of DEC1 was increased in IPF and BLM-injured mice. More importantly, Dec1 -/- mice had reduced PF after BLM challenge. Additionally, DEC1 deficiency relieved EMT development and repressed the PI3K/AKT/GSK-3β/β-catenin integrated signaling pathway in mice and in A549 cells, whereas DEC1 overexpression in vitro had converse effects. Moreover, the PI3K/AKT and Wnt/β-catenin signaling inhibitors, LY294002 and XAV-939, ameliorated BLM-meditated PF in vivo and relieved EMT in vivo and in vitro. These pathways are interconnected by the GSK-3β phosphorylation status. Our findings indicated that during PF progression, DEC1 played a key role in EMT via the PI3K/AKT/GSK-3β/β-catenin integrated signaling pathway. Consequently, targeting DEC1 may be a potential novel therapeutic approach for IPF.
Collapse
Affiliation(s)
- Xingxing Hu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Menglin Zou
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lan Ni
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mingyang Zhang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weishuai Zheng
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bing Liu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhenshun Cheng
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.,Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
41
|
Zhou F, Yao L, Lu X, Li Y, Han X, Wang P. Therapeutic Targeting of GSK3β-Regulated Nrf2 and NFκB Signaling Pathways by Salvianolic Acid A Ameliorates Peritoneal Fibrosis. Front Med (Lausanne) 2022; 9:804899. [PMID: 35321474 PMCID: PMC8936188 DOI: 10.3389/fmed.2022.804899] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/02/2022] [Indexed: 02/06/2023] Open
Abstract
Peritoneal fibrosis is a devastating complication in patients undergoing peritoneal dialysis, with no definite therapy yet available. Salvia miltiorrhiza and its major active component Salvianolic acid A (Sal A) have demonstrated a beneficial effect in myriad diseases. However, their effect on peritoneal fibrosis is unknown. In murine models of peritoneal dialysis, daily Sal A treatment substantially improved the peritoneal dialysis fluid (PDF) elicited peritoneal fibrosis, marked by thickening of the submesothelial compact zone, accumulation of extracellular matrix and increased expression of vimentin and PAI-1, concomitant with attenuation of GSK3β hyperactivity. This coincided with diminished nitrotyrosine in peritoneal tissues and increased Nrf2 nuclear translocation, entailing a lessened oxidative injury and reinforced Nrf2 antioxidant response. Meanwhile, inflammatory infiltration and maladaptive angiogenesis in peritoneal tissues provoked by PDF injury were also mitigated by Sal A, associated with a suppressed NFκB activation. Mechanistically, ectopic expression of the constitutively active GSK3β blunted the NFκB-suppressing and Nrf2-activating efficacy of Sal A in peritoneal mesothelial cells exposed to hypertonic dextrose, suggesting that GSK3β inhibition mediates the protective effect of Sal A. Collectively, our findings may open the avenue for developing a novel therapy based on Sal A for preventing peritoneal fibrosis in peritoneal dialysis.
Collapse
Affiliation(s)
- Fan Zhou
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Molecular Imaging, Zhengzhou, China
| | - Lan Yao
- Blood Purification Center, Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoqing Lu
- Blood Purification Center, Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yubao Li
- Blood Purification Center, Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xingmin Han
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Molecular Imaging, Zhengzhou, China
| | - Pei Wang
- Blood Purification Center, Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Pei Wang
| |
Collapse
|
42
|
Buyuk B, Jin S, Ye K. Epithelial-to-Mesenchymal Transition Signaling Pathways Responsible for Breast Cancer Metastasis. Cell Mol Bioeng 2022; 15:1-13. [PMID: 35096183 PMCID: PMC8761190 DOI: 10.1007/s12195-021-00694-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023] Open
Abstract
Breast carcinoma is highly metastatic and invasive. Tumor metastasis is a convoluted and multistep process involving tumor cell disseminating from their primary site and migrating to the secondary organ. Epithelial-mesenchymal transition (EMT) is one of the crucial steps that initiate cell progression, invasion, and metastasis. During EMT, epithelial cells alter their molecular features and acquire a mesenchymal phenotype. The regulation of EMT is centered by several signaling pathways, including primary mediators TGF-β, Notch, Wnt, TNF-α, Hedgehog, and RTKs. It is also affected by hypoxia and microRNAs (miRNAs). All these pathways are the convergence on the transcriptional factors such as Snail, Slug, Twist, and ZEB1/2. In addition, a line of evidence suggested that EMT and cancer stem like cells (CSCs) are associated. EMT associated cancer stem cells display mesenchymal phenotypes and resist to chemotherapy or targeted therapy. In this review, we highlighted recent discoveries in these signaling pathways and their regulation in breast cancer metastasis and invasion. While the clinical relevance of EMT and breast cancers remains controversial, we speculated a convergent signaling network pivotal to elucidating the transition of epithelial to mesenchymal phenotypes and onset of metastasis of breast cancer cells.
Collapse
Affiliation(s)
- Busra Buyuk
- Department of Biomedical Engineering, Watson College of Engineering and Applied Science, Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), PO Box 6000, Binghamton, NY 13902 USA
| | - Sha Jin
- Department of Biomedical Engineering, Watson College of Engineering and Applied Science, Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), PO Box 6000, Binghamton, NY 13902 USA
| | - Kaiming Ye
- Department of Biomedical Engineering, Watson College of Engineering and Applied Science, Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), PO Box 6000, Binghamton, NY 13902 USA
| |
Collapse
|
43
|
Gibriel AA, Ismail MF, Sleem H, Zayed N, Yosry A, El-Nahaas SM, Shehata NI. Diagnosis and staging of HCV associated fibrosis, cirrhosis and hepatocellular carcinoma with target identification for miR-650, 552-3p, 676-3p, 512-5p and 147b. Cancer Biomark 2022; 34:413-430. [DOI: 10.3233/cbm-210456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND: Chronic HCV infection progresses to fibrosis, cirrhosis and hepatocellular carcinoma (HCC). The latter represents the third most common cause for cancer mortality. Currently, there is no reliable non-invasive biomarker for diagnosis of HCV mediated disorders. OBJECTIVE: Profiling expression signature for circulatory miRNAs in the plasma of 167 Egyptian patients (40 healthy, 48 HCV fibrotic, 39 HCV cirrhotic and 40 HCV-HCC cases). METHODS: QRTPCR was used to quantify expression signature for circulatory miRNAs. RESULTS: MiR-676 and miR-650 were powerful in discriminating cirrhotic and late fibrosis from HCC. MiR-650 could distinguish mild (f0-f1) and advanced (f2-f3) fibrosis from HCC cases. MiR-650 and miR-147b could distinguish early fibrosis from healthy controls meanwhile miR-676 and miR-147b could effectively distinguish between mild chronic and (f1-f3) cases from healthy individuals. All studied miRNAs, except miR-512, can differentiate between (f0-f3) cases and healthy controls. Multivariate logistic regression revealed three potential miRNA panels for effective differentiation of HCC, cirrhotic and chronic liver cases. MiR-676-3p and miR-512-5p were significantly correlated in (f1-f3) fibrosis meanwhile miR-676 and miR-512 could differentiate between cirrhosis and (f0-f3) cases. Both miR-650 and miR-512-5p were positively correlated in the cirrhotic group and in (f0-f4) group. Putative targets for investigated miRNAs were also determined. CONCLUSIONS: Investigated miRNAs could assist in staging and diagnosis of HCV associated disorders.
Collapse
Affiliation(s)
- Abdullah Ahmed Gibriel
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt
| | - Manal Fouad Ismail
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hameis Sleem
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt
| | - Naglaa Zayed
- Endemic Medicine Department and Hepatology Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ayman Yosry
- Endemic Medicine Department and Hepatology Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Saeed M. El-Nahaas
- Endemic Medicine Department and Hepatology Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
44
|
Wen Y, Liu Y, Huang Q, Liu R, Liu J, Zhang F, Liu S, Jiang Y. Moringa oleifera Lam. seed extract protects kidney function in rats with diabetic nephropathy by increasing GSK-3β activity and activating the Nrf2/HO-1 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153856. [PMID: 34856477 DOI: 10.1016/j.phymed.2021.153856] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/26/2021] [Accepted: 11/12/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Moringa oleifera Lam. (M. oleifera) seeds are widely used in traditional folk medicine and as nutritional supplements in the Middle East, Africa, and other regions. Published research showed that M. oleifera seeds (MOS) have pharmacological activities such as blood glucose-lowering, anti-inflammatory, and antitumor effects. However, experimental evidence on the use of MOS to treat diabetic nephropathy and its underlying mechanisms were rarely reported. PURPOSE To evaluate the therapeutic effects of MOS extract on the kidneys of high-fat diet (HFD)-fed rats with streptozotocin-induced diabetic nephropathy and reveal its underlying mechanisms. STUDY DESIGN HFD-fed rats with streptozotocin-induced diabetic nephropathy and high-glucose induced Human Renal Mesangial Cells (HRMC) were used to explore the protective effect of MOS on diabetic nephropathy in vivo and in vitro. METHODS HRMC were used to preliminarily evaluate the effect of MOS extract under high glucose conditions. For the in vivo study, rats were divided into the following 6 groups (n = 5): normal control group (NC), diabetic nephropathy model group (DN), high dose of MOS-treatment group (DN + MOS-H, 200 mg/kg/d); medium dose of MOS-treatment group (DN + MOS-M, 100 mg/kg/d); low dose of MOS-treatment group (DN + MOS-l, 50 mg/kg/d), and metformin-treatment group (DN + MET, 200 mg/kg/d). After 4 weeks of treatment, the damage caused by DN was assessed based on the related parameters of urine and blood. Periodic acid-Schiff (PAS) staining and hematoxylin and eosin (HE) staining were used to assess pathological tissue damage. Immunohistochemistry was used to detect nuclear factor erythroid-derived 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and phosphorylated-glycogen synthase kinase-3beta (P-GSK-3β) levels, whereas western blotting was used to detect Nrf2, HO-1, nephrin, GSK-3β, and p-GSK-3β levels. RESULTS MOS extract could inhibit the proliferation of HRMCs induced by high glucose levels. Compared with the rats in the DN group, MOS not only significantly reduced blood glucose levels and oxidative stress in the experimental rats but also improved their kidney function and reduced kidney tissue damage. Additionally, MOS extract increased GSK-3β activity and the expression of Nrf2 and HO-1. CONCLUSIONS This study showed that MOS could activate GSK-3β and Nrf2/HO-1 pathways to exert antioxidant and anti-renal fibrosis activities, and delay the progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Ying Wen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Yanyang Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Rong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jing Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Fengyu Zhang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Yueping Jiang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
45
|
Peng M, Liu H, Ji Q, Ma P, Niu Y, Ning S, Sun H, Pang X, Yang Y, Zhang Y, Han J, Hao G. Fufang Xueshuantong Improves Diabetic Cardiomyopathy by Regulating the Wnt/ β-Catenin Pathway. Int J Endocrinol 2022; 2022:3919161. [PMID: 36237833 PMCID: PMC9553353 DOI: 10.1155/2022/3919161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/02/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is one of the main complications of diabetic patients and the major reason for the high prevalence of heart failure in diabetic patients. Fufang Xueshuantong (FXST) is a traditional Chinese medicine formula commonly used in the treatment of diabetic retinopathy and stable angina pectoris. However, the role of FXST in DCM has not yet been clarified. This study was conducted to investigate the effects of FXST on diabetic myocardial lesions and reveal its molecular mechanism. The rats were intraperitoneally injected with 65 mg/kg streptozotocin (STZ) to induce diabetes mellitus (DM). DM rats were given saline or FXST. The rats in the control group were intraperitoneally injected with an equal amount of sodium citrate buffer and gavaged with saline. After 12 weeks, echocardiography, heart weight index (HWI), and myocardial pathological changes were determined. The expression of transforming growth factor-beta1 (TGF-β1), collagen I, and collagen III was examined using immunofluorescence staining and western blot. The expressions of Wnt/β-catenin signaling pathway-related proteins and mRNA were detected by western blot and real-time PCR. The results showed that FXST significantly improved cardiac function, ameliorated histopathological changes, and decreased HWI in the DM rats. FXST significantly inhibited the expression of myocardial TGF-β1, collagen I, and collagen III in DM rats. Furthermore, FXST significantly inhibited the Wnt/β-catenin pathway. Taken together, FXST has a protective effect on DCM, which might be mediated by suppressing the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Meizhong Peng
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hanying Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingxuan Ji
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Pan Ma
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yiting Niu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shangqiu Ning
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Huihui Sun
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xinxin Pang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuqian Yang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuting Zhang
- Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Han
- Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Gaimei Hao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Gansu Provincial Hospital of Traditional Chinese Medicine, Gansu, China
| |
Collapse
|
46
|
Elkomy MH, Khallaf RA, Mahmoud MO, Hussein RRS, El-Kalaawy AM, Abdel-Razik ARH, Aboud HM. Intratracheally Inhalable Nifedipine-Loaded Chitosan-PLGA Nanocomposites as a Promising Nanoplatform for Lung Targeting: Snowballed Protection via Regulation of TGF-β/β-Catenin Pathway in Bleomycin-Induced Pulmonary Fibrosis. Pharmaceuticals (Basel) 2021; 14:1225. [PMID: 34959627 PMCID: PMC8707652 DOI: 10.3390/ph14121225] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/16/2022] Open
Abstract
Pulmonary fibrosis is a serious ailment that may progress to lung remodeling and demolition, where the key participants in its incidence are fibroblasts responding to growth factors and cellular calcium swinging. Calcium channel blockers, like nifedipine (NFD), may represent auspicious agents in pulmonary fibrosis treatment. Unfortunately, NFD bears complicated pharmacodynamics and a diminished systemic bioavailability. Thus, the current study aimed to develop a novel, non-invasive nanoplatform for NFD for direct/effective pulmonary targeting via intratracheal instillation. A modified solvent emulsification-evaporation method was adopted for the fabrication of NFD-nanocomposites, integrating poly(D,L-lactide-co-glycolide) (PLGA), chitosan (CTS), and polyvinyl alcohol, and optimized for different physiochemical properties according to the 32 full factorial design. Additionally, the aerodynamic behavior of the nanocomposites was scrutinized through cascade impaction. Moreover, the pharmacokinetic investigations were conducted in rats. Furthermore, the optimum formulation was tested in bleomycin-induced pulmonary fibrosis in rats, wherein fibrotic and oxidative stress parameters were measured. The optimum nanocomposites disclosed a nanosized spherical morphology (226.46 nm), a high entrapment efficiency (61.81%) and a sustained release profile over 24 h (50.4%). As well, it displayed a boosted in vitro lung deposition performance with a mass median aerodynamic diameter of 1.12 µm. Pharmacokinetic studies manifested snowballed bioavailability of the optimal nanocomposites by 3.68- and 2.36-fold compared to both the oral and intratracheal suspensions, respectively. The intratracheal nanocomposites revealed a significant reduction in lung fibrotic and oxidative stress markers notably analogous to normal control besides repairing abnormality in TGF-β/β-catenin pathway. Our results conferred a compelling proof-of-principle that NFD-CTS-PLGA nanocomposites can function as a promising nanoparadigm for pulmonary fibrosis management.
Collapse
Affiliation(s)
- Mohammed H. Elkomy
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt; (R.A.K.); (H.M.A.)
| | - Rasha A. Khallaf
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt; (R.A.K.); (H.M.A.)
| | - Mohamed O. Mahmoud
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt;
| | - Raghda R. S. Hussein
- Department of Clinical Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt;
- Department of Clinical Pharmacy, Faculty of Pharmacy, Modern University for Technology and Information, Cairo 12055, Egypt
| | - Asmaa M. El-Kalaawy
- Department of Pharmacology, Faculty of Medicine, Beni-Suef University, Beni-Suef 62511, Egypt;
| | | | - Heba M. Aboud
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt; (R.A.K.); (H.M.A.)
| |
Collapse
|
47
|
Xu X, Sun X, Wan X, Chen X, Jiang X. Mitomycin induces alveolar epithelial cell senescence by down-regulating GSK3β signaling. Toxicol Lett 2021; 352:61-69. [PMID: 34624459 DOI: 10.1016/j.toxlet.2021.09.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 12/24/2022]
Abstract
Mitomycin treatment induces pulmonary toxicity, and alveolar epithelial cell senescence is crucial in the pathogenesis of the latter. However, the mechanism by which mitomycin induces alveolar epithelial cell senescence has yet to be elucidated. In this work, different doses (37.5-300 nM) of mitomycin induced the senescence of human alveolar type II-like epithelial cells and enhanced the phosphorylation of GSK3β (S9). The GSK3β (S9A) mutant reversed the senescence of mitomycin-treated alveolar epithelial cells. Pharmacological inhibition and gene deletion of Akt1, a kinase that regulates the phosphorylation of GSK3β (S9), suppressed mitomycin-induced alveolar epithelial cell senescence. The knockdown of p53, a downstream effector of GSK3β and an important regulator of cell senescence, repressed mitomycin-induced alveolar epithelial cell senescence. Treatment with baicalein weakened the phosphorylation of GSK3β (S9) and alleviated the senescence of alveolar epithelial cells brought about by mitomycin treatment. GSK3β (S9) phosphorylation appears to be the first signal involved in the mitomycin-induced senescence of alveolar epithelial cells and may present a potential target for attenuating mitomycin-induced pulmonary toxicity.
Collapse
Affiliation(s)
- Xiafang Xu
- College of Pharmaceutical Sciences, Soochow University, China; Shaoxing Maternity and Child Health Care Hospital, China
| | - Xionghua Sun
- College of Pharmaceutical Sciences, Soochow University, China
| | - Xuelei Wan
- College of Pharmaceutical Sciences, Soochow University, China
| | - Xihua Chen
- College of Pharmaceutical Sciences, Soochow University, China
| | - Xiaogang Jiang
- College of Pharmaceutical Sciences, Soochow University, China.
| |
Collapse
|
48
|
Meng X, Liu K, Xie H, Zhu Y, Jin W, Lu J, Wang R. Endoplasmic reticulum stress promotes epithelial‑mesenchymal transition via the PERK signaling pathway in paraquat‑induced pulmonary fibrosis. Mol Med Rep 2021; 24:525. [PMID: 34036384 PMCID: PMC8170262 DOI: 10.3892/mmr.2021.12164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/26/2021] [Indexed: 12/11/2022] Open
Abstract
Pulmonary fibrosis is the primary reason for mortality in patients with paraquat (PQ) poisoning. Our previous study demonstrated that epithelial-mesenchymal transition (EMT) had a role in PQ-induced pulmonary fibrosis. However, the role of endoplasmic reticulum (ER) stress in PQ-induced EMT remains clear. The present study aimed to determine the role of ER stress in EMT in PQ-induced pulmonary fibrosis. A549 and RLE-6TN cells were incubated with LY294002 (a PI3K inhibitor) or transfected with protein kinase RNA-like ER kinase (PERK) small interfering RNA (si) for 24 h prior to being exposed to PQ. Next, the expression levels of ER stress-related proteins, PI3K/AKT/GSK-3β signaling pathway-related proteins and EMT-related markers were analyzed by performing western blotting, reverse transcription-quantitative PCR and immunofluorescence assays. The results of the present study revealed that the protein expression levels of PERK, phosphorylated (p)-PERK, p-eukaryotic initiation factor 2 (eIF2)α were significantly upregulated in the PQ group, whereas p-PI3K, p-AKT and p-GSK-3β were significantly upregulated in the sicontrol + PQ group compared with the sicontrol group. In vitro, following transfection with siPERK or treatment with the PI3K inhibitor, the protein expression levels of E-cadherin (an epithelial marker) were upregulated, whereas the protein expression levels of α-SMA (a mesenchymal marker) were downregulated. Immunofluorescence analysis revealed that the levels of E-cadherin were markedly upregulated, whereas the levels of α-SMA were notably downregulated following transfection with siPERK compared with the sicontrol group. The results of wound healing assay demonstrated that cell migration in the siPERK + PQ group was markedly decreased compared with the sicontrol + PQ group. These indicated that PQ-induced EMT was suppressed after silencing PERK. The expression levels of p-GSK-3β, p-AKT and p-PI3K were also markedly downregulated in the siPERK + PQ group compared with the sicontrol + PQ group. In conclusion, the findings of the present study suggested that ER stress may promote EMT through the PERK signaling pathway in PQ-induced pulmonary fibrosis. Thus, ER stress may represent a potential therapeutic target for PQ-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiaoxiao Meng
- Department of Critical Care Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201620, P.R. China
| | - Kan Liu
- Department of Diving Medicine, Faculty of Nautical Medicine, Second Military Medical University, Shanghai 200082, P.R. China
| | - Hui Xie
- Department of Critical Care Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201620, P.R. China
| | - Yong Zhu
- Department of Critical Care Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201620, P.R. China
| | - Wei Jin
- Department of Critical Care Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201620, P.R. China
| | - Jian Lu
- Department of Critical Care Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201620, P.R. China
| | - Ruilan Wang
- Department of Critical Care Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201620, P.R. China
| |
Collapse
|
49
|
Yu WY, Cai W, Ying HZ, Zhang WY, Zhang HH, Yu CH. Exogenous Plant gma-miR-159a, Identified by miRNA Library Functional Screening, Ameliorated Hepatic Stellate Cell Activation and Inflammation via Inhibiting GSK-3β-Mediated Pathways. J Inflamm Res 2021; 14:2157-2172. [PMID: 34079325 PMCID: PMC8163999 DOI: 10.2147/jir.s304828] [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: 02/04/2021] [Accepted: 04/22/2021] [Indexed: 12/15/2022] Open
Abstract
Purpose Plant-derived exogenous microRNAs (miRNAs) regulate human physiological functions by blocking the translation of target mRNAs. Although several computational approaches have been developed to elucidate the interactions of cross-species miRNAs and their targets in mammals, the number of verified plant miRNAs is still limited, and the biological roles of most exogenous plant miRNAs remain unknown. Methods A miRNA mimic library-based phenotypic screening, which contained 8394 plant mature miRNAs published in the official database miRbase, was performed to identify more novel bioactive plant miRNAs for the prevention of hepatic fibrosis. Inhibition of candidates for the activation of hepatic stellate cells (HSCs) and the underlying mechanisms were evaluated in TGF-β1- and PDGF-exposed HSC models. The protective effects of the candidates against CCl4-induced liver fibrosis were evaluated in a mouse model. Results Among the 8394 plant mature miRNAs reported in the official database miRBase, five candidates were found to effectively inhibit the differentiation of HSCs. gma-miR-159a (miR159a) exerted the strongest inhibitory activities on both TGF-β1- and PDGF-induced HSC activation and proliferation by inhibiting the GSK-3β-mediated NF-κB and TGF-β1 pathways. Moreover, miR159a was mainly accumulated in the liver after intravenous injection, and it reduced CCl4-induced hepatic fibrosis and inflammation in mice. Conclusion Results indicated that miR159a has the therapeutic potential for preventing hepatic fibrosis. This study provides a novel strategy for achieving natural nucleic acid drugs.
Collapse
Affiliation(s)
- Wen-Ying Yu
- Zhejiang Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou, People's Republic of China
| | - Wei Cai
- Department of Traditional Chinese Medicine, Zhejiang Pharmaceutical College, Ningbo, People's Republic of China
| | - Hua-Zhong Ying
- Zhejiang Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou, People's Republic of China
| | - Wen-You Zhang
- Zhejiang Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou, People's Republic of China
| | - Huan-Huan Zhang
- Zhejiang Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou, People's Republic of China
| | - Chen-Huan Yu
- Zhejiang Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou, People's Republic of China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, People's Republic of China.,Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, People's Republic of China
| |
Collapse
|
50
|
Imran SAM, Yazid MD, Idrus RBH, Maarof M, Nordin A, Razali RA, Lokanathan Y. Is There an Interconnection between Epithelial-Mesenchymal Transition (EMT) and Telomere Shortening in Aging? Int J Mol Sci 2021; 22:ijms22083888. [PMID: 33918710 PMCID: PMC8070110 DOI: 10.3390/ijms22083888] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
Epithelial–Mesenchymal Transition (EMT) was first discovered during the transition of cells from the primitive streak during embryogenesis in chicks. It was later discovered that EMT holds greater potential in areas other than the early development of cells and tissues since it also plays a vital role in wound healing and cancer development. EMT can be classified into three types based on physiological functions. EMT type 3, which involves neoplastic development and metastasis, has been the most thoroughly explored. As EMT is often found in cancer stem cells, most research has focused on its association with other factors involving cancer progression, including telomeres. However, as telomeres are also mainly involved in aging, any possible interaction between the two would be worth noting, especially as telomere dysfunction also contributes to cancer and other age-related diseases. Ascertaining the balance between degeneration and cancer development is crucial in cell biology, in which telomeres function as a key regulator between the two extremes. The essential roles that EMT and telomere protection have in aging reveal a potential mutual interaction that has not yet been explored, and which could be used in disease therapy. In this review, the known functions of EMT and telomeres in aging are discussed and their potential interaction in age-related diseases is highlighted.
Collapse
Affiliation(s)
- Siti A. M. Imran
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.A.M.I.); (M.D.Y.); (R.B.H.I.); (M.M.); (A.N.); (R.A.R.)
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.A.M.I.); (M.D.Y.); (R.B.H.I.); (M.M.); (A.N.); (R.A.R.)
| | - Ruszymah Bt Hj Idrus
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.A.M.I.); (M.D.Y.); (R.B.H.I.); (M.M.); (A.N.); (R.A.R.)
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Manira Maarof
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.A.M.I.); (M.D.Y.); (R.B.H.I.); (M.M.); (A.N.); (R.A.R.)
| | - Abid Nordin
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.A.M.I.); (M.D.Y.); (R.B.H.I.); (M.M.); (A.N.); (R.A.R.)
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Rabiatul Adawiyah Razali
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.A.M.I.); (M.D.Y.); (R.B.H.I.); (M.M.); (A.N.); (R.A.R.)
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (S.A.M.I.); (M.D.Y.); (R.B.H.I.); (M.M.); (A.N.); (R.A.R.)
- Correspondence: ; Tel.: +60-391457704
| |
Collapse
|