1
|
Zhao Z, Zeng H, Yu X, Shi Y, Zhao Y, Song Y, Li L, Gao Q, Sun M, Wang B. Hif-1α regulation of the Tet1-β-catenin-Dicer1-miRNAs pathway is involved in depression-like behavior in prenatal hypoxic male offspring. Neuroscience 2025; 576:155-166. [PMID: 40318839 DOI: 10.1016/j.neuroscience.2025.04.051] [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: 12/24/2024] [Revised: 04/22/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Prenatal hypoxia (PH) is a common complication of pregnancy, and it is strongly associated with psychiatric disorders such as depression and anxiety in the offspring. However, how prenatal hypoxia contributes to psychiatric disorders in the offspring is unclear. In this study, we established a model of prenatally hypoxic mice, where pregnant females were treated with hypoxia (10.5% O2) during gestational days 12.5-17.5, while controls (CON) were kept in a normoxic (21% O2) environment. Compared to CON offspring, PH male offspring exhibited depression-like behaviors. Prenatal hypoxia resulted in significantly higher protein level of the oxygen-sensitive subunit of hypoxia-inducible factor (Hif-1α) and lower levels of Ten-eleven translocated methylcytosine dioxygenase 1 (Tet1), β-catenin, and downstream Dicer1-miRNAs pathway associated with depressive behavior. Mechanistically, prenatal hypoxia leads to Hif-1α binding to Tet1, which inhibits β-catenin binding to Tet1, leading to an increase in ubiquitination-dependent degradation of β-catenin and down-regulation of the β-catenin-Dicer1-miRNAs pathway. In addition, administration of the β-catenin-specific agonist SKL2001 or overexpressing virus ameliorated the down-regulation of β-catenin-Dicer1-miRNAs signaling and depression-like behavior in PH male offspring. These findings suggest that Hif-1α and β-catenin competition for Tet1 binding is involved in depression-like behaviors in PH offspring, and this study provides important data on the molecular mechanisms by which prenatal hypoxia might be involved in adult psychiatric disorders of fetal origin.
Collapse
Affiliation(s)
- Zejun Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Hongtao Zeng
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xi Yu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yajun Shi
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yan Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yueyang Song
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lingjun Li
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Qinqin Gao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Miao Sun
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China; McKusick‑Zhang Center for Genetic Medicine, State Key Laboratory for Complex Severe and Rare Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.
| | - Bin Wang
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
2
|
Li C, Cai D, Yuan W, Cai R, Qiu X, Qin Y, Feng Y, Zhu Q, Liu Y, Chen Y, Yuan X, Jiang W, Hou N. The canonical Wnt/β-catenin signaling pathway upregulates carbonic anhydrase 2 via transcription factor 7-like 2 to promote cardiomyopathy in type 2 diabetic mice. Life Sci 2025; 368:123506. [PMID: 40010634 DOI: 10.1016/j.lfs.2025.123506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
Excessive activation of the canonical Wnt/β-catenin pathway contributes to the development of diabetic cardiomyopathy (DCM). Transcription factor 7-like 2 (TCF7L2) is the main β-catenin partner of the TCF family in adult human hearts. Carbonic anhydrase 2 (CA2) is implicated in various hypertrophic cardiomyopathy. In this study, we aimed to investigate the role of the Wnt/β-catenin/TCF7L2 signaling and CA2 in the development of DCM. Streptozotocin (STZ)/high-fat diet (HFD)-induced diabetic mice and high glucose-stimulated neonatal rat cardiomyocytes (NRCMs) were used as in-vivo and in-vitro models of Type 2 diabetes (T2DM), respectively. Histopathological changes in the mouse myocardium were assessed with hematoxylin-eosin (HE) or Masson's trichrome staining. Cardiac function was evaluated with echocardiography. TCF7L2, β-catenin, and CA2 expression was determined with RT-qPCR, western blotting, and immunohistochemistry. Immunoprecipitation (IP) was used to evaluate the formation of the β-catenin/TCF7L2 bipartite. The regulatory relationship between the β-catenin/TCF7L2 bipartite and CA2 was investigated with chromatin immunoprecipitation (ChIP) and a luciferase reporter assay. Compared with the control mice, the T2DM mice exhibited increased myocardial β-catenin and TCF7L2 expression that was concentrated in the nucleus. Treatment of diabetic mice with the β-catenin/TCF7L2 bipartite inhibitor iCRT14 prevented myocardial remodeling and improved cardiac dysfunction. iCRT14 also prevented high glucose-induced hypertrophy in NRCMs, while the β-catenin stabilizer SKL2001 worsened hypertrophy. IP experiments confirmed the formation of the β-catenin/TCF7L2 bipartite in the control and T2DM mouse cardiomyocytes. Moreover, based on the results of RNA-sequencing analysis, CA2 was upregulated in T2DM cardiomyocytes in vitro and in vivo. TCF7L2 overexpression upregulated CA2, while iCRT14 treatment or TCF7L2 knockdown downregulated CA2. CA2 knockdown ameliorated NRCM hypertrophy induced by high glucose and SKL2001. The ChIP experiments revealed an increased interaction between β-catenin/TCF7L2 and the transcription initiation region of CA2 in the heart tissue of T2DM mice. The luciferase reporter assay confirmed that CA2 is directly regulated by the β-catenin/TCF7L2 bipartite. The results indicate that the canonical Wnt/β-catenin pathway upregulates CA2 via TCF7L2 to promote DCM. This research sheds new light on the pathogenesis of DCM and presents new potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Conglin Li
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, PR China; NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China; Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, PR China
| | - Daofeng Cai
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, PR China; NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Wenchang Yuan
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Rui Cai
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Xiaoxia Qiu
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yuan Qin
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yaofeng Feng
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China; KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Qiulian Zhu
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yun Liu
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yilin Chen
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Xun Yuan
- NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Wenyue Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, PR China.
| | - Ning Hou
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, PR China; NMPA Key Laboratory for Clinical Research and Evaluation of Drug for Thoracic Diseases, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
3
|
Ahamad S, Saquib M, Hussain MK, Bhat SA. Targeting Wnt signaling pathway with small-molecule therapeutics for treating osteoporosis. Bioorg Chem 2025; 156:108195. [PMID: 39864370 DOI: 10.1016/j.bioorg.2025.108195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/29/2024] [Accepted: 01/17/2025] [Indexed: 01/28/2025]
Abstract
Small molecules are emerging as potential candidates for treating osteoporosis by activating canonical Wnt signaling. These candidates work either by inhibiting DKK-1, sclerostin, SFRP-1, NOTUM, and S1P lyase or by preventing β-catenin degradation through inhibition of GSK-3β, or by targeting Dvl-CXXC5 and axin/β-catenin interactions. While many of these anti-osteoporotic small molecules are in preclinical development, the paucity of FDA-approved small molecules, or promising candidates, that have progressed to clinical trials for treating bone disorders through this mechanism poses a challenge. Despite advancements in computer-aided drug design, it is rarely employed for designing Wnt signaling activators to treat osteoporosis, and high-throughput screen (HTS) remains the primary method for discovering initial hits. Acknowledging the promising therapeutic potential of these compounds in addressing bone diseases, this review underscores the need for further mechanistic elucidation to enhance our understanding of their applications. Additionally, caution must be exercised in the design of small molecule-based Wnt activators due to their association with oncological risks.
Collapse
Affiliation(s)
- Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University Aligarh 202002 India.
| | - Mohammad Saquib
- Department of Chemistry, University of Allahabad, Prayagraj (Allahabad) 211002, UP, India; Department of Chemistry, G. R. P. B. Degree College, P. R. S. University, Prayagraj (Allahabad) 211010, UP, India
| | | | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
4
|
Jiang Y, Liao C, Lai J, Peng Y, Chen Q, Zheng X. KRT7 promotes pancreatic cancer metastasis by remodeling the extracellular matrix niche through FGF2-fibroblast crosstalk. Sci Rep 2025; 15:6951. [PMID: 40011455 PMCID: PMC11865440 DOI: 10.1038/s41598-024-84129-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 12/20/2024] [Indexed: 02/28/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating cancer with a dismal prognosis due to distant metastasis. Through an analysis of large RNA sequencing and proteomics datasets, we found that high KRT7 expression in PDAC patients was correlated with liver metastasis and poor survival. A functional investigation revealed that the overexpression of KRT7 promoted liver metastasis but did not affect tumor cell proliferation in vivo or in vitro. Analysis of scRNA-Seq data from 24 PDAC samples revealed a negative correlation between KRT7 expression in PDAC cells and cancer-associated fibroblast (CAF) infiltration, and this was further confirmed in orthotopic tumor model mice injected with KRT7-overexpressing PDAC cells, which led the development of to a prometastatic niche with reduced ECM deposition. Mechanistically, KRT7 in PDAC cells promoted the secretion of FGF2, which inhibited CAF proliferation and ECM-related gene transcription through the Wnt/β-catenin pathway. Moreover, targeting FGF2 decreased liver metastasis in vivo. Our study revealed that KRT7 promotes PDAC liver metastasis by remodeling the extracellular matrix niche through FGF2-fibroblast crosstalk and provides a promising strategy for preventing PDAC liver metastasis.
Collapse
Affiliation(s)
- Yuting Jiang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Digestive Endoscopy, Fujian Provincial Hospital, No. 134 East Street, Fuzhou, 350001, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Chengyu Liao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.
- Department of Hepatobiliary Pancreatic Surgery, Fujian Provincial Hospital, No.134 East Street, Fuzhou, 350001, China.
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| | - Jianlin Lai
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Hepatobiliary Pancreatic Surgery, Fujian Provincial Hospital, No.134 East Street, Fuzhou, 350001, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Yunyi Peng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Qilin Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Xiaoling Zheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.
- Department of Digestive Endoscopy, Fujian Provincial Hospital, No. 134 East Street, Fuzhou, 350001, China.
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| |
Collapse
|
5
|
Yan Y, Gong Y, Liang X, Xiong Q, Lin J, Wu Y, Zhang L, Chen H, Jin J, Luan X. Decoding β-catenin associated protein-protein interactions: Emerging cancer therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2025; 1880:189232. [PMID: 39643250 DOI: 10.1016/j.bbcan.2024.189232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
The hyperactive Wnt/β-catenin signaling circuit has been proven to be closely related to the progression of various cancers, with β-catenin serving as a central regulator of pro-tumorigenic processes. Preclinical evidences strongly support β-catenin as a promising therapeutic target. However, it has long been considered "undruggable" due to challenges such as the lack of crystal structures for its N- and C-terminal domains, high mutation rates, and limited availability of inhibitors. Notably, the network of β-catenin-associated protein-protein interactions (PPIs) is vital in the progression of multiple diseases. These interactions form a cancer-specific network that participates in all phases of oncogenesis, from cell metastasis to immunosuppressive microenvironment formation. Thus, researches on these PPIs are essential for unraveling the molecular mechanisms behind tumors with aberrant β-catenin activation, as well as for developing new targeted therapies. In this review, we delve into how β-catenin's PPIs orchestrate cancer progression and highlight biological and clinical dilemmas, proposing frontier technologies and potential challenges in targeting β-catenin for cancer therapy.
Collapse
Affiliation(s)
- Yue Yan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yiting Gong
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaohui Liang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qingyi Xiong
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiayi Lin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ye Wu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lijun Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jinmei Jin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
6
|
Castagnoli L, Franceschini A, Cancila V, Dugo M, Bigliardi M, Chiodoni C, Toneguzzo P, Regondi V, Corsetto PA, Pietrantonio F, Mazzucchelli S, Corsi F, Belfiore A, Vingiani A, Pruneri G, Ligorio F, Colombo MP, Tagliabue E, Tripodo C, Vernieri C, Triulzi T, Pupa SM. CD36 enrichment in HER2-positive mesenchymal stem cells drives therapy refractoriness in breast cancer. J Exp Clin Cancer Res 2025; 44:19. [PMID: 39833955 PMCID: PMC11744895 DOI: 10.1186/s13046-025-03276-z] [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: 03/27/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Growing evidence shows that the reprogramming of fatty acid (FA) metabolism plays a key role in HER2-positive (HER2 +) breast cancer (BC) aggressiveness, therapy resistance and cancer stemness. In particular, HER2 + BC has been defined as a "lipogenic disease" due to the functional and bi-directional crosstalk occurring between HER2-mediated oncogenic signaling and FA biosynthesis via FA synthase activity. In this context, the functional role exerted by the reprogramming of CD36-mediated FA uptake in HER2 + BC poor prognosis and therapy resistance remains unclear. In this study, we aimed to elucidate whether enhanced CD36 in mesenchymal HER2 + cancer stem cells (CSCs) is directly involved in anti-HER2 treatment refractoriness in HER2 + BC and to design future metabolism-based approaches targeting both FA reprogramming and the "root" of cancer. METHODS Molecular, biological and functional characterization of CD36-mediated FA uptake was investigated in HER2 + BC patients, cell lines, epithelial and mesenchymal CSCs. Cell proliferation was analyzed by SRB assay upon treatment with lapatinib, CD36 inhibitor, or Wnt antagonist/agonist. Engineered cell models were generated via lentivirus infection and transient silencing. CSC-like properties and tumorigenesis of HER2 + BC cells with or without CD36 depletion were examined by mammosphere forming efficiency assay, flow cytometry, cell sorting, ALDH activity assay and xenograft mouse model. FA uptake was examined by flow cytometry with FA BODIPY FL C16. Intratumor expression of CSC subsets was evaluated via multiplex immunostaining and immunolocalization analysis. RESULTS Molecular data demonstrated that CD36 is significantly upmodulated on treatment in therapy resistant HER2 + BC patients and its expression levels in BC cells is correlated with FA uptake. We provided evidence of a consistent enrichment of CD36 in HER2 + epithelial-mesenchymal transition (EMT)-like CSCs from all tested resistant cell models that mechanistically occurs via Wnt signaling pathway activation. Consistently, both in vitro and in vivo dual blockade of CD36 and HER2 increased the anti-CSC efficacy of anti-HER2 drugs favoring the transition of the therapy resistant mesenchymal CSCs into therapy-sensitive mesenchymal-epithelial transition (MET)-like epithelial state. In addition, expression of CD36 in intratumor HER2 + mesenchymal CSCs is significantly associated with resistance to trastuzumab in HER2 + BC patients. CONCLUSIONS These results support the metabolo-oncogenic nature of CD36-mediated FA uptake in HER2 + therapy-refractory BC. Our study provides evidence that targeting CD36 might be an effective metabolic therapeutic strategy in the treatment of this malignancy.
Collapse
Affiliation(s)
- Lorenzo Castagnoli
- Microenvironment and Biomarkers of Solid Tumors Unit, Department of Experimental Oncology, Amadeolab Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Alma Franceschini
- Microenvironment and Biomarkers of Solid Tumors Unit, Department of Experimental Oncology, Amadeolab Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department PROMISE, Universita' Di Palermo, Palermo, Italy
| | - Matteo Dugo
- Breast Cancer Unit Clinical Translational and Immunotherapy Research, Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Martina Bigliardi
- Microenvironment and Biomarkers of Solid Tumors Unit, Department of Experimental Oncology, Amadeolab Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Claudia Chiodoni
- Molecular Immunology Unit, Department of Experimental Oncology, Amadeolab Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Paolo Toneguzzo
- Microenvironment and Biomarkers of Solid Tumors Unit, Department of Experimental Oncology, Amadeolab Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Viola Regondi
- Microenvironment and Biomarkers of Solid Tumors Unit, Department of Experimental Oncology, Amadeolab Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Paola A Corsetto
- Department of Pharmacologicaland, Biomolecular Sciences "Rodolfo Paoletti", Università Di Milano, Milan, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Serena Mazzucchelli
- Department of Biomedical and Clinical Sciences, Università Di Milano, Milan, Italy
| | - Fabio Corsi
- Surgery Department, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Antonio Belfiore
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Antonio Vingiani
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
- Department of Oncology and Hemato-Oncology, Universita' di Milano, Milan, Italy
| | - Giancarlo Pruneri
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
- Department of Oncology and Hemato-Oncology, Universita' di Milano, Milan, Italy
| | - Francesca Ligorio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Mario P Colombo
- Molecular Immunology Unit, Department of Experimental Oncology, Amadeolab Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Elda Tagliabue
- Microenvironment and Biomarkers of Solid Tumors Unit, Department of Experimental Oncology, Amadeolab Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department PROMISE, Universita' Di Palermo, Palermo, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Tiziana Triulzi
- Microenvironment and Biomarkers of Solid Tumors Unit, Department of Experimental Oncology, Amadeolab Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Serenella M Pupa
- Microenvironment and Biomarkers of Solid Tumors Unit, Department of Experimental Oncology, Amadeolab Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy.
| |
Collapse
|
7
|
Din ZU, Cui B, Wang C, Zhang X, Mehmood A, Peng F, Liu Q. Crosstalk between lipid metabolism and EMT: emerging mechanisms and cancer therapy. Mol Cell Biochem 2025; 480:103-118. [PMID: 38622439 DOI: 10.1007/s11010-024-04995-1] [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: 11/18/2023] [Accepted: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Lipids are the key component of all membranes composed of a variety of molecules that transduce intracellular signaling and provide energy to the cells in the absence of nutrients. Alteration in lipid metabolism is a major factor for cancer heterogeneity and a newly identified cancer hallmark. Reprogramming of lipid metabolism affects the diverse cancer phenotypes, especially epithelial-mesenchymal transition (EMT). EMT activation is considered to be an essential step for tumor metastasis, which exhibits a crucial role in the biological processes including development, wound healing, and stem cell maintenance, and has been widely reported to contribute pathologically to cancer progression. Altered lipid metabolism triggers EMT and activates multiple EMT-associated oncogenic pathways. Although the role of lipid metabolism-induced EMT in tumorigenesis is an attractive field of research, there are still significant gaps in understanding the underlying mechanisms and the precise contributions of this interplay. Further study is needed to clarify the specific molecular mechanisms driving the crosstalk between lipid metabolism and EMT, as well as to determine the potential therapeutic implications. The increased dependency of tumor cells on lipid metabolism represents a novel therapeutic target, and targeting altered lipid metabolism holds promise as a strategy to suppress EMT and ultimately inhibit metastasis.
Collapse
Affiliation(s)
- Zaheer Ud Din
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, Guangdong Medical University, Dongguan, China
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Cenxin Wang
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
| | - Xiaoyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
| | - Arshad Mehmood
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China.
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
8
|
Mousavi SM, Jalali-Zefrei F, Shourmij M, Tabaghi S, Davari A, Khalili SB, Farzipour S, Salari A. Targeting Wnt Pathways with Small Molecules as New Approach in Cardiovascular Disease. Curr Cardiol Rev 2025; 21:108-122. [PMID: 39482911 PMCID: PMC12060913 DOI: 10.2174/011573403x333038241023153349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 11/03/2024] Open
Abstract
The increasing incidences of morbidity and mortality associated with cardiovascular diseases represent significant difficulties for clinical treatment and have a major impact on patient health. Wnt signaling pathways are highly conserved and are well known for their regulatory roles in embryonic development, tissue regeneration, and adult tissue homeostasis. Wnt signaling is classified into two distinct pathways: canonical Wnt/β-catenin signaling and noncanonical pathways, including planar cell polarity and Wnt/Ca2+ pathways. A growing body of experimental evidence suggests the involvement of both canonical and non-canonical Wnt signaling pathways in the development of cardiovascular diseases, including myocardial hypertrophy, arrhythmias, diabetic cardiomyopathy, arrhythmogenic cardiomyopathy, and myocardial infarction. Thus, to enhance patient quality of life, diagnosing and treating cardiac illnesses may require a thorough understanding of the molecular functions played by the Wnt pathway in these disorders. Many small-molecule inhibitors specifically target various components within the Wnt signaling pathways, such as Frizzled, Disheveled, Porcupine, and Tankyrase. This study aims to present an overview of the latest findings regarding the functions of Wnt signaling in human cardiac disorders and possible inhibitors of Wnt, which could lead to novel approaches for treating cardiac ailments.
Collapse
Affiliation(s)
- Seyed Mehdi Mousavi
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Jalali-Zefrei
- Department of radiology, Faculty of Medicine, Guilan University of Medical Science, Rasht, Iran
| | - Mohammad Shourmij
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Shiva Tabaghi
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhesam Davari
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeed Bahador Khalili
- Department of Electronic Engineering, Universitat Rovira i Virgili, Tarragona, 43007, Spain
| | - Soghra Farzipour
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
9
|
Xu L, Liu B, Ma H, Qi E, Ma J, Chang T, Zhang J, Zhang W, Chen W, Cao X, Xiong X. O-GlcNAc transferase promotes vascular smooth muscle calcification through modulating Wnt/β-catenin signaling. FASEB J 2024; 38:e70271. [PMID: 39704274 DOI: 10.1096/fj.202401649rr] [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: 07/17/2024] [Revised: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
Vascular calcification (VC), associated with high cardiovascular mortality in patients with chronic kidney disease (CKD), involves osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs). O-GlcNAcylation, a dynamic post-translational modification, is closely linked to cardiovascular diseases, including VC. However, the exact role and molecular mechanism of O-GlcNAc signaling in abnormal mineral metabolism-induced VC remain unclear. In the current study, we found that the levels of O-GlcNAc transferase (OGT) and global protein O-GlcNAcylation were significantly upregulated in the artery tissues of mouse calcification models and CKD patients with VC. To further delineate the in vivo role of OGT in VC, we generated Ogt smooth muscle cell-specific knockout mice and challenged them with 5/6 nephrectomy (5/6 Nx) or high-dose vitamin D3 to induce VC. Deletion of Ogt in VSMCs led to alleviated VC in response to 5/6 Nx or VD3. Moreover, elevated O-GlcNAcylation, induced by Thiamet-G, facilitated osteogenic transdifferentiation in VSMCs in response to phosphate, whereas OSMI-1, which reduces O-GlcNAcylation, exhibited an opposite phenotypic effect. Mechanistically, O-GlcNAc signaling enhanced the osteogenic conversion of VSMCs through regulation of canonical Wnt/β-catenin pathway. Indeed, β-catenin was O-GlcNAcylated by OGT and further increased its transcriptional activity in VSMCs. Furthermore, pharmacological activation of Wnt/β-catenin signaling largely reversed the diminished aortic calcification caused by Ogt ablation. Our findings demonstrate that smooth muscle O-GlcNAc signaling plays an important role in regulating hyperphosphatemia-induced VC and reveal that O-GlcNAcylation of β-catenin protein modulates its content and activity in VSMCs.
Collapse
MESH Headings
- Animals
- N-Acetylglucosaminyltransferases/metabolism
- N-Acetylglucosaminyltransferases/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Wnt Signaling Pathway
- Mice
- Mice, Knockout
- Humans
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Male
- Mice, Inbred C57BL
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- beta Catenin/metabolism
- Cells, Cultured
- Osteogenesis
Collapse
Affiliation(s)
- Lin Xu
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Boao Liu
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Honghui Ma
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Enbo Qi
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jie Ma
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Tingmin Chang
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Jinghong Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wencheng Zhang
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Weiqian Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Xuan Cao
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Xiwen Xiong
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
10
|
Yi Y, Che W, Xu P, Mao C, Li Z, Wang Q, Lyu J, Wang X. Conversion of glioma cells into neuron-like cells by small molecules. iScience 2024; 27:111091. [PMID: 39483145 PMCID: PMC11525470 DOI: 10.1016/j.isci.2024.111091] [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: 03/12/2024] [Revised: 06/25/2024] [Accepted: 09/29/2024] [Indexed: 11/03/2024] Open
Abstract
Currently, researchers are exploring the conversion of astrocytes into functional mature neurons and gradually exploring the conversion of glioma into neurons. We report that SLCDS (SB431542, LDN193189, CHIR99021, DAPT, and SKL2001) has been shown to convert human glioma cells into mature neuron-like cells. The converted cells exhibited upregulation of DCX, TuJ1, MAP2, NeuN, and GAD67, while the expressions of EGFR, PDGFR, Ki67, and vimentin were inhibited. The nTFs, such as NeuroD1 and Sox2, were upregulated, along with TF genes associated with neurogenesis and tumor suppression. We have finally confirmed that overexpressing nTFs can induce the conversion of glioma cells into neuronal cells. This study demonstrates that SLCDS can activate the expression of nTFs in human glioma cells and induce the conversion of human glioma cells into neuron-like cells. Additionally, SLCDS inhibits the expressions of EGFR, PDGFR, Ki67, and Vimentin in gliomas. Our findings offer a potential approach for treating glioma.
Collapse
Affiliation(s)
- Yongjun Yi
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou 510632, P.R. China
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, P.R. China
| | - Wenqiang Che
- Department of Neurosurgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, P.R. China
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, P.R. China
| | - Ping Xu
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou 510630, P.R. China
| | - Chuxiao Mao
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou 510630, P.R. China
| | - Zhizhong Li
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou 510630, P.R. China
| | - Qingsong Wang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou 510630, P.R. China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou 510632, P.R. China
| | - Xiangyu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, P.R. China
| |
Collapse
|
11
|
Zheng J, Yang H, Liu C, Zhang R, Yibulayimu N, Jin X. Ethanol Extract of Anacyclus pyrethrum Root Ameliorates Cough-Variant Asthma Through the TLR4/NF-κB Pathway and Wnt/β-Catenin Pathway. Mol Biotechnol 2024; 66:3274-3284. [PMID: 37910337 DOI: 10.1007/s12033-023-00935-4] [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: 04/02/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023]
Abstract
Cough-variant asthma (CVA) has been recognized as the initial stage or pre-asthmatic state of classic asthma, which characterized by cough as the primary clinical presentation. Inhaled glucocorticoids, oral leukotriene receptor antagonists and antihistamines are the clinical treatments, but their efficacy is not satisfactory. Some traditional Chinese medicine (TCM) has been reported to have certain advantages in the treatment of CVA, but the underlying molecular mechanisms are still unclear. Recent research has indicated that Anacyclus pyerhrurm (L) DC. is commonly used in the treatment of human diseases. The aim of our study was to evaluate the anti-inflammatory and anti-oxidative mechanism of the ethanol extract of Anacyclus pyrethrum (L) DC. root (EEAP) in a model of CVA. In our study, we indicated that EEAP ameliorated CVA by reducing cough frequency and inflammatory effect and oxidative stress in an in vivo rat model of CVA. In addition, EEAP ameliorated LPS-induced cell apoptosis and regulated inflammatory effect and oxidative stress in vitro. Mechanistically, EEAP exerted anti-inflammatory effects through regulating the TLR4/NF-κB pathway and Wnt/β-catenin pathway, and overexpressing TLR4 or activating the Wnt/β-catenin pathway by SKL2001 reversed EEAP-exerted effects in LPS-exposed BEAS-2B and 16-HBE cells. In conclusion, EEAP attenuated cell apoptosis, inflammation and oxidative stress through restraining the TLR4/NF-κB pathway and Wnt/β-catenin pathway in CVA, which shown that EEAP might be a promising therapeutic agent for CVA and may provide a theoretical basis for clinical treatment with CVA patients.
Collapse
Affiliation(s)
- Jun Zheng
- Department of Critical Care Medicine, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hao Yang
- Department of Pharmacy, The Sixth Affiliated Hospital of Xinjiang Medical University, No. 39, Wuxing South Road, TianShan District, Urumqi, 830000, China
| | - Changjiang Liu
- Department of Pharmacy, The Sixth Affiliated Hospital of Xinjiang Medical University, No. 39, Wuxing South Road, TianShan District, Urumqi, 830000, China
| | - Rui Zhang
- Department of Pharmacy, The Sixth Affiliated Hospital of Xinjiang Medical University, No. 39, Wuxing South Road, TianShan District, Urumqi, 830000, China
| | - Nadire Yibulayimu
- Market Supervision and Administration Bureau of Huocheng County, HuoCheng, Ili, China
| | - Xiaoyue Jin
- Department of Pharmacy, The Sixth Affiliated Hospital of Xinjiang Medical University, No. 39, Wuxing South Road, TianShan District, Urumqi, 830000, China.
| |
Collapse
|
12
|
Lan T, Quan W, Yu DH, Chen X, Wang ZF, Li ZQ. High expression of LncRNA HOTAIR is a risk factor for temozolomide resistance in glioblastoma via activation of the miR-214/β-catenin/MGMT pathway. Sci Rep 2024; 14:26224. [PMID: 39482401 PMCID: PMC11528118 DOI: 10.1038/s41598-024-77348-z] [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: 07/02/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
HOX transcript antisense RNA (HOTAIR) is upregulated in glioblastoma (GBM) and associated with temozolomide (TMZ) resistance. However, the mechanisms underlying HOTAIR-mediated TMZ resistance remains poorly understood. HOTAIR expression in glioma-related public datasets and drug response estimation were analyzed using bioinformatics. These findings were verified by overexpressing HOTAIR in TMZ-sensitive U251 cells and/or silencing HOTAIR in resistant U251 cells (U251R). The cytotoxic effects were evaluated using cell viability assay and flow cytometry analysis of cell cycle and apoptosis. In this study, we found that HOTAIR was upregulated in TMZ-resistant GBM cell lines and patients with high HOTAIR expression responded poorly to TMZ therapy. HOTAIR knockdown restored TMZ sensitivity in U251R cells, while HOTAIR overexpression conferred TMZ resistance in U251 cells. Wnt/β-catenin signaling was enriched in patients with high HOTAIR expression; consistently, HOTAIR positively regulated β-catenin expression in U251 cells. Moreover, HOTAIR-mediated TMZ resistance was associated with increased MGMT protein level, which resulted from the HOTAIR/miR-214-3p/β-catenin network. Besides, GBM with high HOTAIR expression exhibited sensitivity to methotrexate. Methotrexate enhanced TMZ sensitivity in U251R cells, accompanied by reduced expression of HOTAIR and β-catenin. Thus, we conlcude that HOTAIR is a risk factor for TMZ resistance and methotrexate may represent a potential therapeutic drug for patients with high HOTAIR expression level.
Collapse
Affiliation(s)
- Tian Lan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Quan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dong-Hu Yu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xi Chen
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ze-Fen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, China.
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
13
|
Zhou S, Qian K, Yu S, Zhao Y, Shen Q, Li Y. MiR-4429 Alleviates Malignant Behaviors of Lung Adenocarcioma Through Wnt/β-Catenin Pathway. Cancer Biother Radiopharm 2024; 39:562-572. [PMID: 34491827 DOI: 10.1089/cbr.2021.0154] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Lung adenocarcinoma (ADC) is a common subtype of non-small cell lung cancer. MicroRNAs have been reported to be effective biomarkers for diagnosis and an important target for therapy. MiR-4429 is a newly identified miRNA, which can take part in tumor progression as a tumor inhibitor. Moreover, it is an exosomal miRNA that can be taken by lung ADC cell line A549. Nevertheless, its role in lung ADC has been poorly studied. This research discovered that miR-4429 was low expressed in lung ADC cells. MiR-4429 mimics could alleviate the capacities of cell proliferation and metastasis. The mimics are able to reverse epithelial-mesenchymal transition at the same time. Furthermore, it was verified that miR-4429 could bind to β-catenin and negatively regulate β-catenin expression. Interestingly, SKL2001 can reverse the role of miR-4429 on tumor. Consequently, miR-4429 can inactivate Wnt/β-catenin signaling pathway by targeting β-catenin and prevent oncogene expression in lung ADC cells.
Collapse
Affiliation(s)
- Shaoqiang Zhou
- Department of Breast Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Kebao Qian
- Department of Thoracic Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shuhui Yu
- Department of Radiation Therapy Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yutao Zhao
- Department of Radiation Therapy Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qin Shen
- Institute of Neuroscience, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan Province, China
| | - Ya Li
- Department of Radiation Therapy Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
14
|
Yuan M, Shi H, Wang B, Cai J, Yu W, Wang W, Qian Q, Wang Y, Zhou X, Liu J. Targeting SOCS2 alleviates myocardial fibrosis by reducing nuclear translocation of β-catenin. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119804. [PMID: 39084528 DOI: 10.1016/j.bbamcr.2024.119804] [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: 03/13/2024] [Revised: 06/25/2024] [Accepted: 07/20/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Myocardial fibrosis is an important pathological feature of dilated cardiomyopathy (DCM). The roles of SOCS2 in fibrosis of different organs are controversial. Herein, we investigated the function and potential mechanism of SOCS2 in myocardial fibrosis. METHODS Bioinformatics, immunohistochemistry (IHC), immunofluorescence (IF), western blot (WB), real-time fluorescence quantitative PCR (qPCR), rat primary myocardial fibroblasts (rCFs) culture, doxorubicin (DOX) induced mouse dilated cardiomyopathy (DCM) model, and in vivo adeno-associated virus (AAV) infection were used to explore the role of SOCS2 in DCM. RESULTS Bioinformatics analysis showed that SOCS2 was positively correlated with fibrosis related factors. SOCS2 was significantly upregulated in patients and mice with DCM. In vivo experiments showed that targeted inhibition of cardiac SOCS2 could improve mouse cardiac function and alleviate myocardial fibrosis. Further research demonstrated that SOCS2 promoted the transformation of myofibroblasts. Knockdown of SOCS2 reduced the nuclear localization of β-catenin, which inhibited the fibrogenic effect of Wnt/β-catenin pathway. In addition, bioinformatics analysis suggested that lymphoid enhancer binding factor 1 (LEF1) was significantly positively correlated with SOCS2. Finally, dual luciferase assays demonstrated that LEF1 could bind to the promoter region of SOCS2, thereby mediating its transcriptional activation. CONCLUSION SOCS2 could activate the Wnt/β-catenin by regulating the nuclear translocation of β-catenin, which induces the transcriptional activation of SOCS2. Overall, these results indicated a positive feedback activation phenomenon between SOCS2, β-catenin and LEF1 in DCM. These results suggested that inhibition of SOCS2 could effectively alleviate the progression of myocardial fibrosis and improve cardiac function.
Collapse
Affiliation(s)
- Ming Yuan
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Hongjie Shi
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Bin Wang
- Department of Cardiovascular Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Jie Cai
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Wenjun Yu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Wei Wang
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Qiaofeng Qian
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Yumou Wang
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China
| | - Xianwu Zhou
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China.
| | - Jinping Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China; Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan 430071, China.
| |
Collapse
|
15
|
Dennison NR, Fusenig M, Grönnert L, Maitz MF, Ramirez Martinez MA, Wobus M, Freudenberg U, Bornhäuser M, Friedrichs J, Westenskow PD, Werner C. Precision Culture Scaling to Establish High-Throughput Vasculogenesis Models. Adv Healthc Mater 2024; 13:e2400388. [PMID: 38465502 DOI: 10.1002/adhm.202400388] [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: 02/21/2024] [Indexed: 03/12/2024]
Abstract
Hydrogel-based 3D cell cultures can recapitulate (patho)physiological phenomena ex vivo. However, due to their complex multifactorial regulation, adapting these tissue and disease models for high-throughput screening workflows remains challenging. In this study, a new precision culture scaling (PCS-X) methodology combines statistical techniques (design of experiment and multiple linear regression) with automated, parallelized experiments and analyses to customize hydrogel-based vasculogenesis cultures using human umbilical vein endothelial cells and retinal microvascular endothelial cells. Variations of cell density, growth factor supplementation, and media composition are systematically explored to induce vasculogenesis in endothelial mono- and cocultures with mesenchymal stromal cells or retinal microvascular pericytes in 384-well plate formats. The developed cultures are shown to respond to vasculogenesis inhibitors in a compound- and dose-dependent manner, demonstrating the scope and power of PCS-X in creating parallelized tissue and disease models for drug discovery and individualized therapies.
Collapse
Affiliation(s)
- Nicholas R Dennison
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
| | - Maximilian Fusenig
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
- Medical Clinic I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Lisa Grönnert
- Ocular Technologies, Immunology, Infectious Diseases and Ophthalmology, Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Manfred F Maitz
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
| | | | - Manja Wobus
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
| | - Uwe Freudenberg
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
| | - Martin Bornhäuser
- Medical Clinic I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Jens Friedrichs
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
| | - Peter D Westenskow
- Ocular Technologies, Immunology, Infectious Diseases and Ophthalmology, Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, 01069, Dresden, Germany
- Medical Clinic I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
- Center for Regenerative Therapies Dresden and Cluster of Excellence Physics of Life, Technische Universität Dresden, 01307, Dresden, Germany
| |
Collapse
|
16
|
Zhu M, Han Y, Gu T, Wang R, Si X, Kong D, Zhao P, Wang X, Li J, Zhai X, Yu Z, Lu H, Li J, Huang H, Qian P. Class I HDAC inhibitors enhance antitumor efficacy and persistence of CAR-T cells by activation of the Wnt pathway. Cell Rep 2024; 43:114065. [PMID: 38578828 DOI: 10.1016/j.celrep.2024.114065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/18/2024] [Accepted: 03/21/2024] [Indexed: 04/07/2024] Open
Abstract
Epigenetic modification shapes differentiation trajectory and regulates the exhaustion state of chimeric antigen receptor T (CAR-T) cells. Limited efficacy induced by terminal exhaustion closely ties with intrinsic transcriptional regulation. However, the comprehensive regulatory mechanisms remain largely elusive. Here, we identify class I histone deacetylase inhibitors (HDACi) as boosters of CAR-T cell function by high-throughput screening of chromatin-modifying drugs, in which M344 and chidamide enhance memory maintenance and resistance to exhaustion of CAR-T cells that induce sustained antitumor efficacy both in vitro and in vivo. Mechanistically, HDACi decrease HDAC1 expression and enhance H3K27ac activity. Multi-omics analyses from RNA-seq, ATAC-seq, and H3K27ac CUT&Tag-seq show that HDACi upregulate expression of TCF4, LEF1, and CTNNB1, which subsequently activate the canonical Wnt/β-catenin pathway. Collectively, our findings elucidate the functional roles of class I HDACi in enhancing CAR-T cell function, which provides the basis and therapeutic targets for synergic combination of CAR-T cell therapy and HDACi treatment.
Collapse
Affiliation(s)
- Meng Zhu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Yingli Han
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Tianning Gu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China; Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Rui Wang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Xiaohui Si
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China; Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Delin Kong
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China; Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Zhao
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Xiujian Wang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China; Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinxin Li
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Xingyuan Zhai
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China; Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zebin Yu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Huan Lu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Jingyi Li
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - He Huang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China; Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Pengxu Qian
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University & Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China.
| |
Collapse
|
17
|
Li Y, Luan S, Ruan C, Li W, Zhang X, Ran Z, Bi W, Tong Y, Gao L, Zhao J, Li Y, He Z. TSHR signaling promotes hippocampal dependent memory formation through modulating Wnt5a/β-catenin mediated neurogenesis. Biochem Biophys Res Commun 2024; 704:149723. [PMID: 38430698 DOI: 10.1016/j.bbrc.2024.149723] [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: 12/11/2023] [Revised: 02/14/2024] [Accepted: 02/24/2024] [Indexed: 03/05/2024]
Abstract
Subclinical hyperthyroidism is defined biochemically as a low or undetectable thyroid-stimulating hormone (TSH) with normal thyroid hormone levels. Low TSHR signaling is considered to associate with cognitive impairment. However, the underlying molecular mechanism by which TSHR signaling modulates memory is poorly understood. In this study, we found that Tshr-deficient in the hippocampal neurons impairs the learning and memory abilities of mice, accompanying by a decline in the number of newborn neurons. Notably, Tshr ablation in the hippocampus decreases the expression of Wnt5a, thereby inactivating the β-catenin signaling pathway to reduce the neurogenesis. Conversely, activating of the Wnt/β-catenin pathway by the agonist SKL2001 results in an increase in hippocampal neurogenesis, resulting in the amelioration in the deficits of memory caused by Tshr deletion. Understanding how TSHR signaling in the hippocampus regulates memory provides insights into subclinical hyperthyroidism affecting cognitive function and will suggest ways to rationally design interventions for neurocognitive disorders.
Collapse
Affiliation(s)
- Yuchen Li
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Sisi Luan
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China; Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China
| | - Cairong Ruan
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Weihao Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Xinyu Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Zijing Ran
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Wenkai Bi
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Yuelin Tong
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Ling Gao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Jiajun Zhao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
| | - Yuan Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
| | - Zhao He
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
| |
Collapse
|
18
|
Xu Z, Elaish M, Wong CP, Hassan BB, Lopez-Orozco J, Felix-Lopez A, Ogando NS, Nagata L, Mahal LK, Kumar A, Wilson JA, Noyce R, Mayers I, Power C, Evans D, Hobman TC. The Wnt/β-catenin pathway is important for replication of SARS-CoV-2 and other pathogenic RNA viruses. NPJ VIRUSES 2024; 2:6. [PMID: 40295745 PMCID: PMC11721380 DOI: 10.1038/s44298-024-00018-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/10/2024] [Indexed: 04/30/2025]
Abstract
Understanding how viruses affect cellular pathways during infection may facilitate development of host cell-targeted therapeutics with broad-spectrum antiviral activity. The interferon (IFN) response is critical for reducing replication and pathogenesis of many viruses including Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19. Mounting evidence indicates that peroxisomes which are best known as metabolic organelles, function in the IFN response. Recently, we reported that the Wnt/β-catenin signaling pathway strongly suppresses peroxisome biogenesis. Here, we show that SARS-CoV-2 infection activates Wnt/β-catenin signaling and hypothesized that pharmacological inhibition of this pathway would result in increased peroxisome formation and enhanced IFN production. Indeed, Wnt/β-catenin signaling potently inhibits replication of SARS-CoV-2 and other pathogenic RNA viruses in vitro and reduces viral load, inflammation and clinical symptoms in a mouse model of COVID-19. As such, targeting this cellular pathway may have prophylactic and/or therapeutic value in reducing the disease burden caused by emerging viral pathogens.
Collapse
Affiliation(s)
- Zaikun Xu
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | - Mohamed Elaish
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Cheung Pang Wong
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada
| | - Bardes B Hassan
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | | | - Alberto Felix-Lopez
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada
| | - Natacha S Ogando
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Les Nagata
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada
| | - Lara K Mahal
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Anil Kumar
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Joyce A Wilson
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ryan Noyce
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Irv Mayers
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Christopher Power
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - David Evans
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Tom C Hobman
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada.
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
19
|
Qiu L, Sun Y, Ning H, Chen G, Zhao W, Gao Y. The scaffold protein AXIN1: gene ontology, signal network, and physiological function. Cell Commun Signal 2024; 22:77. [PMID: 38291457 PMCID: PMC10826278 DOI: 10.1186/s12964-024-01482-4] [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] [Accepted: 01/06/2024] [Indexed: 02/01/2024] Open
Abstract
AXIN1, has been initially identified as a prominent antagonist within the WNT/β-catenin signaling pathway, and subsequently unveiled its integral involvement across a diverse spectrum of signaling cascades. These encompass the WNT/β-catenin, Hippo, TGFβ, AMPK, mTOR, MAPK, and antioxidant signaling pathways. The versatile engagement of AXIN1 underscores its pivotal role in the modulation of developmental biological signaling, maintenance of metabolic homeostasis, and coordination of cellular stress responses. The multifaceted functionalities of AXIN1 render it as a compelling candidate for targeted intervention in the realms of degenerative pathologies, systemic metabolic disorders, cancer therapeutics, and anti-aging strategies. This review provides an intricate exploration of the mechanisms governing mammalian AXIN1 gene expression and protein turnover since its initial discovery, while also elucidating its significance in the regulation of signaling pathways, tissue development, and carcinogenesis. Furthermore, we have introduced the innovative concept of the AXIN1-Associated Phosphokinase Complex (AAPC), where the scaffold protein AXIN1 assumes a pivotal role in orchestrating site-specific phosphorylation modifications through interactions with various phosphokinases and their respective substrates.
Collapse
Affiliation(s)
- Lu Qiu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yixuan Sun
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Haoming Ning
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Guanyu Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Wenshan Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
20
|
Lin X, Meng X, Lin J. The possible role of Wnt/β-catenin signalling in vitiligo treatment. J Eur Acad Dermatol Venereol 2023; 37:2208-2221. [PMID: 36912722 DOI: 10.1111/jdv.19022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 02/08/2023] [Indexed: 03/14/2023]
Abstract
Vitiligo is a common chronic skin disease which has an adverse impact on patients' life. Its pathogenesis is complex, involving autoimmunity and oxidative stress (OS). Autoimmunity leads to the loss of epidermal melanocytes and the formation of the depigmented patches of the disease. Treatment of vitiligo should control the exaggerated immune response to arrest the progress of active disease, and then promote melanocytes to repigmentation. Wnt/β-catenin signalling pathway has been of recent interest in vitiligo. Wnt/β-catenin signalling pathway is downregulated in vitiligo. Upregulation of Wnt/β-catenin signalling possibly control vitiligo autoimmune response by protecting melanocyte from OS damage, inhibiting CD8+ T cell effector cell differentiation and enhancing Treg. Wnt/β-catenin signalling plays a critical role in the melanocyte regeneration by driving the differentiation of melanocyte stem cells (McSCs) into melanocytes. Promoting Wnt/β-catenin signalling can not only arrest the progress of active disease of vitiligo but also promote repigmentation. Some of the main effective therapies for vitiligo are likely to work by activating Wnt/β-catenin signalling. Agents that can enhance the effect of Wnt/β-catenin signalling may become potential candidates for the development of new drugs for vitiligo treatment.
Collapse
Affiliation(s)
- Xiran Lin
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xianmin Meng
- Department of Pathology and Laboratory Medicine, Axia Women's Health, Oaks, Pennsylvania, USA
| | - Jingrong Lin
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
21
|
Boccazzi M, Macchiarulo G, Lebon S, Janowska J, Le Charpentier T, Faivre V, Hua J, Marangon D, Lecca D, Fumagalli M, Mani S, Abbracchio MP, Gressens P, Schang AL, Van Steenwinckel J. G protein-coupled receptor 17 is regulated by WNT pathway during oligodendrocyte precursor cell differentiation. Neurobiol Dis 2023; 187:106315. [PMID: 37783234 DOI: 10.1016/j.nbd.2023.106315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/13/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023] Open
Abstract
G protein-coupled receptor 17 (GPR17) and the WNT pathway are critical players of oligodendrocyte (OL) differentiation acting as essential timers in developing brain to achieve fully-myelinating cells. However, whether and how these two systems are related to each other is still unknown. Of interest, both factors are dysregulated in developing and adult brain diseases, including white matter injury and cancer, making the understanding of their reciprocal interactions of potential importance for identifying new targets and strategies for myelin repair. Here, by a combined pharmacological and biotechnological approach, we examined regulatory mechanisms linking WNT signaling to GPR17 expression in OLs. We first analyzed the relative expression of mRNAs encoding for GPR17 and the T cell factor/Lymphoid enhancer-binding factor-1 (TCF/LEF) transcription factors of the canonical WNT/β-CATENIN pathway, in PDGFRα+ and O4+ OLs during mouse post-natal development. In O4+ cells, Gpr17 mRNA level peaked at post-natal day 14 and then decreased concomitantly to the physiological uprise of WNT tone, as shown by increased Lef1 mRNA level. The link between WNT signaling and GPR17 expression was further reinforced in vitro in primary PDGFRα+ cells and in Oli-neu cells. High WNT tone impaired OL differentiation and drastically reduced GPR17 mRNA and protein levels. In Oli-neu cells, WNT/β-CATENIN activation repressed Gpr17 promoter activity through both putative WNT response elements (WRE) and upregulation of the inhibitor of DNA-binding protein 2 (Id2). We conclude that the WNT pathway influences OL maturation by repressing GPR17, which could have implications in pathologies characterized by dysregulations of the OL lineage including multiple sclerosis and oligodendroglioma.
Collapse
Affiliation(s)
- Marta Boccazzi
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | | | - Sophie Lebon
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Justyna Janowska
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | | | - Valérie Faivre
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Jennifer Hua
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, 20133 Milan, Italy
| | - Shyamala Mani
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France
| | - Anne-Laure Schang
- Université Paris Cité, Inserm, NeuroDiderot, F-75019 Paris, France; Université Paris Cité, UMR 1153 CRESS, Paris, France.
| | | |
Collapse
|
22
|
Li Y, Yao L, Lu J. IL-35 inhibits adipogenesis via PPARγ-Wnt/β-catenin signaling pathway by targeting Axin2. Int Immunopharmacol 2023; 122:110615. [PMID: 37429144 DOI: 10.1016/j.intimp.2023.110615] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/24/2023] [Accepted: 07/04/2023] [Indexed: 07/12/2023]
Abstract
Interleukin (IL)-35, a member of the IL-12 family, functions as an immunosuppressive cytokine that plays a crucial role in the regulation of immune-related disorders and inflammatory diseases. Adipose tissue, which is now recognized as an immune organ, is regulated by immunocytes through various signaling pathways, including the peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα) pathway and the Wnt/β-actin pathway. However, there is limited research regarding the effects of IL-35 on adipogenesis. Our current findings indicated that IL-35 impedes the proliferation and promotes the cytotoxicity of 3T3-L1 preadipocytes. Furthermore, IL-35 inhibited the adipogenic differentiation, as well as suppressed triglyceride and lipid accumulation. Additionally, the expression of PPARγ and C/EBPα, two key regulators of adipogenesis, were both down-regulated with IL-35 treatment. In order to explicate the mechanisms underlying the effects of IL-35, we conducted an investigation into the expression of Axin2, an intracellular inhibitor of Wnt/β-catenin signaling, in 3T3-L1 preadipocyte cells. Gene silencing of Axin2 through small interfering RNAs (siRNAs) enhanced PPARγ and C/EBPα expression while decreasing nuclear β-catenin levels in the presence of IL-35. Furthermore, in IL-35-treated cells, Axin2 knockdown boosted adipogenic differentiation (as measured by increased Oil Red O staining). These findings imply that IL-35 regulates Axin2 expression and thereby plays an important role in adipocyte development.
Collapse
Affiliation(s)
- Yuxuan Li
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, No. 36 San Hao Street, Heping District, Shenyang, 110004, PR China
| | - Lutian Yao
- Department of Orthopedics, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang 110001, PR China.
| | - Jing Lu
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, PR China.
| |
Collapse
|
23
|
Tang S, Leng M, Tan C, Zhu L, Pang Y, Zhang X, Chang YF, Lin W. Critical role for ribonucleoside-diphosphate reductase subunit M2 in ALV-J-induced activation of Wnt/β-catenin signaling via interaction with P27. J Virol 2023; 97:e0026723. [PMID: 37582207 PMCID: PMC10506463 DOI: 10.1128/jvi.00267-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/20/2023] [Indexed: 08/17/2023] Open
Abstract
Avian leukemia virus subgroup J (ALV-J) causes various diseases associated with tumor formation and decreased fertility and induced immunosuppressive disease, resulting in significant economic losses in the poultry industry globally. Virus usually exploits the host cellular machinery for their replication. Although there are increasing evidences for the cellular proteins involving viral replication, the interaction between ALV-J and host proteins leading to the pivotal steps of viral life cycle are still unclear. Here, we reported that ribonucleoside-diphosphate reductase subunit M2 (RRM2) plays a critical role during ALV-J infection by interacting with capsid protein P27 and activating Wnt/β-catenin signaling. We found that the expression of RRM2 is effectively increased during ALV-J infection, and that RRM2 facilitates ALV-J replication by interacting with viral capsid protein P27. Furthermore, ALV-J P27 activated Wnt/β-catenin signaling by promoting β-catenin entry into the nucleus, and RRM2 activated Wnt/β-catenin signaling by enhancing its phosphorylation at Ser18 during ALV-J infection. These data suggest that the upregulation of RRM2 expression by ALV-J infection favors viral replication in host cells via activating Wnt/β-catenin signaling. IMPORTANCE Our results revealed a novel mechanism by which RRM2 facilitates ALV-J growth. That is, the upregulation of RRM2 expression by ALV-J infection favors viral replication by interacting with capsid protein P27 and activating Wnt/β-catenin pathway in host cells. Furthermore, the phosphorylation of serine at position 18 of RRM2 was verified to be the important factor regulating the activation of Wnt/β-catenin signaling. This study provides insights for further studies of the molecular mechanism of ALV-J infection.
Collapse
Affiliation(s)
- Shuang Tang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Provincial Animal Virus Vector Vaccine Engineering Technology Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Mei Leng
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Provincial Animal Virus Vector Vaccine Engineering Technology Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Chen Tan
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Provincial Animal Virus Vector Vaccine Engineering Technology Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Lin Zhu
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Provincial Animal Virus Vector Vaccine Engineering Technology Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yanling Pang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Provincial Animal Virus Vector Vaccine Engineering Technology Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xinheng Zhang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Provincial Animal Virus Vector Vaccine Engineering Technology Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Wencheng Lin
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangdong Provincial Animal Virus Vector Vaccine Engineering Technology Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
24
|
An L, Gong H, Yu X, Zhang W, Liu X, Yang X, Shu L, Liu J, Yang L. Downregulation of MAL2 inhibits breast cancer progression through regulating β-catenin/c-Myc axis. Cancer Cell Int 2023; 23:144. [PMID: 37480012 PMCID: PMC10362617 DOI: 10.1186/s12935-023-02993-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/13/2023] [Indexed: 07/23/2023] Open
Abstract
PURPOSE Myelin and lymphocyte protein 2 (MAL2) is mainly involved in endocytosis under physiological conditions and mediates the transport of materials across the membranes of cell and organelle. It has been reported that MAL2 is significantly upregulated in diverse cancers. This study aimed to investigate the role of MAL2 in breast cancer (BC). METHODS Bioinformatics analysis and Immunohistochemical assay were applied to detect the correlation between MAL2 expression in breast cancer tissues and the prognosis of breast cancer patients. Functional experiments were carried out to investigate the role of MAL2 in vitro and in vivo. The molecular mechanisms involved in MAL2-induced β-catenin and c-Myc expression and β-catenin/c-Myc-mediated enhancement of BC progression were confirmed by western blot, β-catenin inhibitor and agonist, Co-IP and immunofluorescence colocalization assays. RESULTS Results from the cancer genome atlas (TCGA) and clinical samples confirmed a significant upregulation of MAL2 in BC tissues than in adjacent non-tumor tissues. High expression of MAL2 was associated with worse prognosis. Functional experiments demonstrated that MAL2 knockdown reduced the migration and invasion associating with EMT, increased the apoptosis of BC cells in vitro and reduced the metastatic capacity in vivo. Mechanistically, MAL2 interacts with β-catenin in BC cells. MAL2 silencing reduced the expression of β-catenin and c-Myc, while the β-catenin agonist SKL2001 partially rescued the downregulation of c-Myc and inhibition of migration and invasion caused by MAL2 knockdown in BC cells. CONCLUSION These observations provided evidence that MAL2 acted as a potential tumor promoter by regulating EMT and β-catenin/c-Myc axis, suggesting potential implications for anti-metastatic therapy for BC.
Collapse
Affiliation(s)
- Lijun An
- Department of Immunology, Basic Medical College, Guizhou Medical University, Dongqing Road, Guian New District, Guiyang, Guizhou, 550004, China
| | - Huiyuan Gong
- Department of Immunology, Basic Medical College, Guizhou Medical University, Dongqing Road, Guian New District, Guiyang, Guizhou, 550004, China
| | - Xiaojing Yu
- Department of Immunology, Basic Medical College, Guizhou Medical University, Dongqing Road, Guian New District, Guiyang, Guizhou, 550004, China
| | - Wangming Zhang
- Department of Immunology, Basic Medical College, Guizhou Medical University, Dongqing Road, Guian New District, Guiyang, Guizhou, 550004, China
| | - Xiaohua Liu
- Department of Immunology, Basic Medical College, Guizhou Medical University, Dongqing Road, Guian New District, Guiyang, Guizhou, 550004, China
| | - Xiaomin Yang
- Department of Immunology, Basic Medical College, Guizhou Medical University, Dongqing Road, Guian New District, Guiyang, Guizhou, 550004, China
| | - Liping Shu
- Department of Immunology, Basic Medical College, Guizhou Medical University, Dongqing Road, Guian New District, Guiyang, Guizhou, 550004, China
| | - Jielin Liu
- Department of Immunology, Basic Medical College, Guizhou Medical University, Dongqing Road, Guian New District, Guiyang, Guizhou, 550004, China
| | - Liuqi Yang
- Department of Immunology, Basic Medical College, Guizhou Medical University, Dongqing Road, Guian New District, Guiyang, Guizhou, 550004, China.
| |
Collapse
|
25
|
Yan Z, Ruan B, Wang S, Du T, Shao X, Chen G, Wang L, Zhai D, Zhu S, Lu Z, Cao X. RNA-binding Protein QKI Inhibits Osteogenic Differentiation Via Suppressing Wnt Pathway. Arch Med Res 2023; 54:102853. [PMID: 37460362 DOI: 10.1016/j.arcmed.2023.102853] [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: 01/25/2023] [Revised: 05/23/2023] [Accepted: 07/03/2023] [Indexed: 07/31/2023]
Abstract
BACKGROUND Dysregulation of MSCs differentiation is associated with many pathophysiological processes. Genetically modified MSCs transplantation helps restore bone loss efficiently. METHODS BMSCs-specific QKI overexpressing and knockdown mice were built to explore QKI's role in bone formation and fat accumulation. Primary BMSCs with QKI overexpression and knockout were subjected to osteogenic and adipogenic differentiation. ALP staining and oil red O staining were performed to evaluate the differences between the groups. RNA immunoprecipitation was performed to identify the QKI-related pathway. QKI deficient BMSCs were transplanted into mice with glucocorticoid-induced osteoporosis to evaluate its therapeutic potential. RESULTS Mice harboring BMSC-specific transgenic QKI exhibited reduced bone mass, while BMSC-specific QKI-deficient mice showed an increase in bone mass. Osteogenic differentiation of QKI deficient BMSCs was promoted and adipogenic differentiation was inhibited, while QKI overexpression in BMSCs displayed the opposite effects. To define the underlying mechanisms, RIP sequencing was performed. Wnt pathway-related genes were the putative direct target mRNAs of QKI, Canonical Wnt pathway activation was involved in QKI's effects on osteogenic differentiation. RNA immunoprecipitation quantitative real-time Polymerase Chain Reaction (PCR) and RNA fluorescence in situ hybridization experiments further validated that QKI repressed the expressions of Wnt5b, Fzd7, Dvl3 and β-catenin via direct binding to their putative mRNA specific sites. Glucocorticoid-induced osteoporotic mice transplanted with QKI deficient BMSCs exhibited less bone loss compared with mice transplanted with control BMSCs. CONCLUSIONS QKI suppressed BMSCs osteogenic differentiation by downregulating the expressions of Wnt5b, Fzd7, Dvl3 and β-catenin. Loss of QKI in BMSCs transplantation may provide a new strategy for the treatment of orthopedic diseases such as osteoporosis.
Collapse
Affiliation(s)
- Zhao Yan
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China; Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Banjun Ruan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shan Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tianshu Du
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaolong Shao
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guo Chen
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, China
| | - Li Wang
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, China
| | - Dongsheng Zhai
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, China
| | - Shu Zhu
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zifan Lu
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, Fourth Military Medical University, Xi'an, China; Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xiaorui Cao
- PLA Institute of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
26
|
Deng X, Zeng Y, Qiu X, Zhong M, Xiong X, Luo M, Zhang J, Chen X. CRIP1 supports the growth and migration of AML-M5 subtype cells by activating Wnt/β-catenin pathway. Leuk Res 2023; 130:107312. [PMID: 37224580 DOI: 10.1016/j.leukres.2023.107312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/26/2023]
Abstract
Acute myeloid leukemia (AML) is a clinically and molecularly heterogeneous hematopoietic disorder. To effectively eradicate AML, it is urgent to develop new therapeutic approaches and identify novel molecular targets. In silico analysis indicated that the expression of cysteine-rich intestinal protein 1 (CRIP1) was significantly elevated in AML cells and correlated with worse overall survival of the AML patients. However, its specific roles in AML remain elusive. Here we demonstrated that CRIP1 acted as a key oncogene to support AML cell survival and migration. Using a loss-of-function analysis, we found that CRIP1 silencing in U937 and THP1 cells by lentivirus-mediated shRNAs resulted in a decrease in cell growth, migration and colony formation, and an increase in chemosensitivity to Ara-C. CRIP1 silencing induced cell apoptosis and G1/S transition arrest. Mechanically, CRIP1 silencing caused inactivation of Wnt/β-catenin pathway through upregulating axin1 protein. The Wnt/β-catenin agonist SKL2001 markedly rescued the cell growth and migration defect induced by CRIP1 silencing. Our findings reveals that CRIP1 may contribute to AML-M5 pathogenesis and represent a novel target for AML-M5 treatment.
Collapse
Affiliation(s)
- Xiaoling Deng
- Jiangxi Health Commission Key Laboratory of Leukemia, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China; Ganzhou Key Laboratory of Molecular Medicine, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China
| | - Yanmei Zeng
- Jiangxi Health Commission Key Laboratory of Leukemia, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China; Ganzhou Key Laboratory of Molecular Medicine, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China
| | - Xiaofen Qiu
- Jiangxi Health Commission Key Laboratory of Leukemia, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China; Ganzhou Key Laboratory of Molecular Medicine, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China
| | - Mingxing Zhong
- Jiangxi Health Commission Key Laboratory of Leukemia, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China
| | - Xiujuan Xiong
- Department of Pathology, Basic Medical College of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Mansheng Luo
- Clinical laboratory, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China
| | - Jingdong Zhang
- Jiangxi Health Commission Key Laboratory of Leukemia, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China
| | - Xiaoli Chen
- Jiangxi Health Commission Key Laboratory of Leukemia, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China; Ganzhou Key Laboratory of Molecular Medicine, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China.
| |
Collapse
|
27
|
Wang J, Zhao D, Lei Z, Ge P, Lu Z, Chai Q, Zhang Y, Qiang L, Yu Y, Zhang X, Li B, Zhu S, Zhang L, Liu CH. TRIM27 maintains gut homeostasis by promoting intestinal stem cell self-renewal. Cell Mol Immunol 2023; 20:158-174. [PMID: 36596873 PMCID: PMC9887071 DOI: 10.1038/s41423-022-00963-1] [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: 08/06/2022] [Accepted: 11/20/2022] [Indexed: 01/05/2023] Open
Abstract
Dysregulation of gut homeostasis is associated with irritable bowel syndrome (IBS), a chronic functional gastrointestinal disorder affecting approximately 11.2% of the global population. The poorly understood pathogenesis of IBS has impeded its treatment. Here, we report that the E3 ubiquitin ligase tripartite motif-containing 27 (TRIM27) is weakly expressed in IBS but highly expressed in inflammatory bowel disease (IBD), a frequent chronic organic gastrointestinal disorder. Accordingly, knockout of Trim27 in mice causes spontaneously occurring IBS-like symptoms, including increased visceral hyperalgesia and abnormal stool features, as observed in IBS patients. Mechanistically, TRIM27 stabilizes β-catenin and thus activates Wnt/β-catenin signaling to promote intestinal stem cell (ISC) self-renewal. Consistent with these findings, Trim27 deficiency disrupts organoid formation, which is rescued by reintroducing TRIM27 or β-catenin. Furthermore, Wnt/β-catenin signaling activator treatment ameliorates IBS symptoms by promoting ISC self-renewal. Taken together, these data indicate that TRIM27 is critical for maintaining gut homeostasis, suggesting that targeting the TRIM27/Wnt/β-catenin axis could be a potential treatment strategy for IBS. Our study also indicates that TRIM27 might serve as a potential biomarker for differentiating IBS from IBD.
Collapse
Affiliation(s)
- Jing Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Dongdong Zhao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Zehui Lei
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Pupu Ge
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhe Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Qiyao Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yong Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Lihua Qiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Yang Yu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Xinwen Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bingxi Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shu Zhu
- Institute of Immunology, Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, 100850, China.
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China.
| |
Collapse
|
28
|
Wang S, Hu T, He M, Gu Y, Cao X, Yuan Z, Lv X, Getachew T, Quan K, Sun W. Defining ovine dermal papilla cell markers and identifying key signaling pathways regulating its intrinsic properties. Front Vet Sci 2023; 10:1127501. [PMID: 36923053 PMCID: PMC10009177 DOI: 10.3389/fvets.2023.1127501] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/09/2023] [Indexed: 03/02/2023] Open
Abstract
Dermal papilla cell (DPC), one of the key cell types during hair follicle development and regeneration, specifies hair size, shape and cycling. It is also an important in vitro screening model for hair growth. Although some characteristics of DPCs, such as agglutinative growth and marker genes, have been studied in mice and humans, the intrinsic properties of ovine DPCs and the regulatory mechanism of the intrinsic properties during continued culture in vitro remained unknown. In this study, based on our previous single-cell transcriptome sequencing on sheep lambskin, we verified SOX18 and PDGFRA as the novel marker genes of ovine DPCs through immunofluorescence staining on skin sections and cultured DPCs. Using continued cell culture and alkaline phosphatase staining, we found that different from mice and humans, ovine DPCs exhibit particularly robust and stable aggregation with unbated alkaline phosphatase activity till 30 passages during continued culture in vitro. Also, we found that the expression of some marker genes and the activity of Wnt/β-catenin signaling differ between early passaged DPCs and multiple passaged DPCs. Further, using Wnt/β-catenin agonist and antagonist, we demonstrated that Wnt/β-catenin signaling could regulate cell aggregation and alkaline phosphatase activity of ovine DPCs through regulating FGF and IGF signaling. This study provides the basis for isolating ovine DPCs and defines their intrinsic properties, which contribute to improving wool performance and medicine of hair regeneration.
Collapse
Affiliation(s)
- Shanhe Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Tingyan Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Mingliang He
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Yifei Gu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Xiukai Cao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China.,International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou, China
| | - Zehu Yuan
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China.,International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou, China
| | - Xiaoyang Lv
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China.,International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou, China
| | - Tesfaye Getachew
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa, Ethiopia
| | - Kai Quan
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Wei Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China.,International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou, China.,"Innovative China" "Belt and Road" International Agricultural Technology Innovation Institute for Evaluation, Protection, and Improvement on Sheep Genetic Resource, Yangzhou, China
| |
Collapse
|
29
|
Hu T, Lv X, Getachew T, Mwacharo JM, Haile A, Quan K, Li Y, Wang S, Sun W. Effect of Sox18 on the Induction Ability of Dermal Papilla Cells in Hu Sheep. BIOLOGY 2022; 12:biology12010065. [PMID: 36671756 PMCID: PMC9855062 DOI: 10.3390/biology12010065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022]
Abstract
Sox18 is a developmental gene that encodes transcription factors. It has been indicated as be a key gene affecting the growth and development of hair follicles, in which dermal papilla cells (DPCs) have been demonstrated to play an important role through their ability to induce the formation of hair follicles. Pre-laboratory studies have found that Sox18 is differentially expressed in the dermal papilla cells of different pattern types of Hu sheep. We speculated that Sox18 plays an important role in the dermal papilla cells of Hu sheep. In our study, we analyzed the effect of Sox18 on the induction ability of DPCs in order to elucidate the function and molecular mechanism of Sox18 in the DPCs of Hu sheep. We first identified the expression of Sox18 in the DPCs of Hu sheep by immunofluorescence staining. We then used alkaline phosphatase staining, cell morphology observations and RT-PCR to detect the effect of Sox18 on the induction of DPCs after overexpression of or interference with Sox18. We also used RT-PCR, WB and immunofluorescence staining to detect the effect of Sox18 on the Wnt/β-catenin signal pathway in DPCs. We found that Sox18 was specifically expressed in the DPCs of Hu sheep, and that Sox18 could enhance the alkaline phosphatase activity in the DPCs of Hu sheep and accelerate cell agglutination. The results of RT-PCR revealed that Sox18 promoted the mRNA expression of Versican, HHIP and FGFRI, and inhibited the mRNA expression of BMP4 and WIF1. Further studies showed that Sox18 promoted the expression of β-catenin and activated the Wnt/β-catenin signal pathway in DPCs. When the Wnt/β-catenin signal pathway of DPCs was activated, the induction ability of DPCs was enhanced. Overall, we believe that Sox18 could enhance the induction ability of DPCs in Hu sheep and regulate the induction ability of DPCs through the Wnt/β-catenin signal pathway.
Collapse
Affiliation(s)
- Tingyan Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou 225009, China
| | - Xiaoyang Lv
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Tesfaye Getachew
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa 999047, Ethiopia
| | - Joram M. Mwacharo
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa 999047, Ethiopia
| | - Aynalem Haile
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa 999047, Ethiopia
| | - Kai Quan
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou 450060, China
| | - Yutao Li
- CSIRO Agriculture and Food, 306 Carmody Rd, St Lucia 4067, Australia
| | - Shanhe Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou 225009, China
- Correspondence: (S.W.); (W.S.)
| | - Wei Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- “Innovative China” “Belt and Road” International Agricultural Technology Innovation Institute for Evaluation, Protection, and Improvement on Sheep Genetic Resource, Yangzhou University, Yangzhou 225009, China
- Correspondence: (S.W.); (W.S.)
| |
Collapse
|
30
|
Hu Z, Ma J, Yue H, Luo Y, Li X, Wang C, Wang L, Sun B, Chen Z, Wang L, Gu Y. Involvement of LIN28A in Wnt-dependent regulation of hippocampal neurogenesis in the aging brain. Stem Cell Reports 2022; 17:1666-1682. [PMID: 35750042 PMCID: PMC9287676 DOI: 10.1016/j.stemcr.2022.05.016] [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: 01/07/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 10/29/2022] Open
Abstract
Hippocampal neurogenesis declines with aging. Wnt ligands and antagonists within the hippocampal neurogenic niche regulate the proliferation of neural progenitor cells and the development of new neurons, and the changes of their levels in the niche mediate aging-associated decline of neurogenesis. We found that RNA-binding protein LIN28A remained existent in neural progenitor cells and granule neurons in the adult hippocampus and that it decreased with aging. Lin28a knockout inhibited the responsiveness of neural progenitor cells to niche Wnt agonists and reduced neurogenesis, thus impairing pattern separation. Overexpression of Lin28a increased the proliferation of neural progenitor cells, promoted the functional integration of newborn neurons, restored neurogenesis in Wnt-deficient dentate gyrus, and rescued the impaired pattern separation in aging mice. Our data suggest that LIN28A regulates adult hippocampal neurogenesis as an intracellular mechanism by responding to niche Wnt signals, and its decrease is involved in aging-associated decline of hippocampal neurogenesis and related cognitive functions.
Collapse
Affiliation(s)
- Zhechun Hu
- Center of Stem Cell and Regenerative Medicine and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jiao Ma
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Huimin Yue
- Center of Stem Cell and Regenerative Medicine and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yujian Luo
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Xiaofang Li
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Chao Wang
- Center of Stem Cell and Regenerative Medicine and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Liang Wang
- Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital, Mental Health Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Binggui Sun
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China; MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310058, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Lang Wang
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou 310029, China.
| | - Yan Gu
- Center of Stem Cell and Regenerative Medicine and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou 310058, China; MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
31
|
Kadian LK, Arora M, Prasad CP, Pramanik R, Chauhan SS. Signaling pathways and their potential therapeutic utility in esophageal squamous cell carcinoma. Clin Transl Oncol 2022; 24:1014-1032. [PMID: 34990001 DOI: 10.1007/s12094-021-02763-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022]
Abstract
Esophageal cancer is a complex gastrointestinal malignancy with an extremely poor outcome. Approximately 80% of cases of this malignancy in Asian countries including India are of squamous cell origin, termed Esophageal Squamous Cell Carcinoma (ESCC).The five-year survival rate in ESCC patients is less than 20%. Neo-adjuvant chemo-radiotherapy (NACRT) followed by surgical resection remains the major therapeutic strategy for patients with operable ESCC. However, resistance to NACRT and local recurrence after initial treatment are the leading cause of dismal outcomes in these patients. Therefore, an alternative strategy to promote response to the therapy and reduce the post-operative disease recurrence is highly needed. At the molecular level, wide variations have been observed in tumor characteristics among different populations, nevertheless, several common molecular features have been identified which orchestrate disease progression and clinical outcome in the malignancy. Therefore, determination of candidate molecular pathways for targeted therapy remains the mainstream idea of focus in ESCC research. In this review, we have discussed the key signaling pathways associated with ESCC, i.e., Notch, Wnt, and Nrf2 pathways, and their crosstalk during disease progression. We further discuss the recent developments of novel agents to target these pathways in the context of targeted cancer therapy. In-depth research of the signaling pathways, gene signatures, and a combinatorial approach may help in discovering targeted therapy for ESCC.
Collapse
Affiliation(s)
- L K Kadian
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - M Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - C P Prasad
- Department of Medical Oncology (Lab), Dr. B. R. Ambedkar-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - R Pramanik
- Department of Medical Oncology, Dr. B. R. Ambedkar-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - S S Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
32
|
Xu Z, Wang C, He Y, Mao X, Zhang MZ, Hou YP, Li B. Hypoxia-Inducible Factor Protects Against Acute Kidney Injury via the Wnt/β-Catenin Signaling Pathway. Am J Physiol Renal Physiol 2022; 322:F611-F624. [PMID: 35403451 DOI: 10.1152/ajprenal.00023.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Promoting adaptive repair (AR) in acute kidney injury (AKI) is an effective strategy to preventprogression from AKI to chronic kidney disease (CKD). However, the mechanisms involved in renal repair after AKI remain unclear. In this study, we investigated the role of hypoxia-inducible factor (HIF), an important regulator of ischemic and hypoxic injury, in AKI during the repair phase. We established mouse models of ischemia-reperfusion injury (IRI)-induced AKI with AR or maladaptive repair (MAR). We found that after injury, the activation of HIF in the AR group was rapid, while in the MAR group, HIF activation was relatively delayed, and its expression was significantly lower than that in the AR group during the early repair phase. To further investigate the mechanism of HIF, we regulated the expression of HIF-1α and HIF-2α in HK-2 cells and EA.hy926 cells, respectively. Silencing HIF expression reduced proliferation and increased apoptosis in cells injured by hypoxia/reoxygenation (H/R). Self-healing ability was further reduced due to the downregulation of HIF. Moreover, HIF overexpression had the opposite effect. HIF increased the expression of β-catenin and its downstream target genes. Activation of Wnt/β-catenin by the small molecule activator SKL2001 mitigated the damaging effect of HIF knockdown, while blocking β-catenin with the inhibitor IWR-1-endo reduced the protective effects of HIF. In conclusion, HIF, which is highly expressed in the early stage after AKI, promotes renal repair by interacting with the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- ZhiHui Xu
- Department of Nephrology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chang Wang
- Department of Nephrology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, China
| | - YiXin He
- Department of Nephrology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - XinYue Mao
- Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Man-Zhu Zhang
- Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yan-Pei Hou
- Department of Nephrology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bing Li
- Department of Nephrology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
33
|
Chen X, Li Y, Yuan X, Yuan W, Li C, Zeng Y, Lian Y, Qiu X, Qin Y, Zhang G, Liu X, Luo C, Luo JD, Hou N. Methazolamide Attenuates the Development of Diabetic Cardiomyopathy by Promoting β-Catenin Degradation in Type 1 Diabetic Mice. Diabetes 2022; 71:795-811. [PMID: 35043173 DOI: 10.2337/db21-0506] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022]
Abstract
Methazolamide (MTZ), a carbonic anhydrase inhibitor, has been shown to inhibit cardiomyocyte hypertrophy and exert a hypoglycemic effect in patients with type 2 diabetes and diabetic db/db mice. However, whether MTZ has a cardioprotective effect in the setting of diabetic cardiomyopathy is not clear. We investigated the effects of MTZ in a mouse model of streptozotocin-induced type 1 diabetes mellitus (T1DM). Diabetic mice received MTZ by intragastric gavage (10, 25, or 50 mg/kg, daily for 16 weeks). In the diabetic group, MTZ significantly reduced both random and fasting blood glucose levels and improved glucose tolerance in a dose-dependent manner. MTZ ameliorated T1DM-induced changes in cardiac morphology and dysfunction. Mechanistic analysis revealed that MTZ blunted T1DM-induced enhanced expression of β-catenin. Similar results were observed in neonatal rat cardiomyocytes (NRCMs) and adult mouse cardiomyocytes treated with high glucose or Wnt3a (a β-catenin activator). There was no significant change in β-catenin mRNA levels in cardiac tissues or NRCMs. MTZ-mediated β-catenin downregulation was recovered by MG132, a proteasome inhibitor. Immunoprecipitation and immunofluorescence analyses showed augmentation of AXIN1-β-catenin interaction by MTZ in T1DM hearts and in NRCMs treated with Wnt3a; thus, MTZ may potentiate AXIN1-β-catenin linkage to increase β-catenin degradation. Overall, MTZ may alleviate cardiac hypertrophy by mediating AXIN1-β-catenin interaction to promote degradation and inhibition of β-catenin activity. These findings may help inform novel therapeutic strategy to prevent heart failure in patients with diabetes.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yilang Li
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xun Yuan
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Wenchang Yuan
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Conglin Li
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yue Zeng
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yuling Lian
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiaoxia Qiu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Zhujiang Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Yuan Qin
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Guiping Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiawen Liu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Chengfeng Luo
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jian-Dong Luo
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ning Hou
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
34
|
Liu Y, Ruan X, Li J, Wang B, Chen J, Wang X, Wang P, Tu X. The Osteocyte Stimulated by Wnt Agonist SKL2001 Is a Safe Osteogenic Niche Improving Bioactivities in a Polycaprolactone and Cell Integrated 3D Module. Cells 2022; 11:cells11050831. [PMID: 35269452 PMCID: PMC8909416 DOI: 10.3390/cells11050831] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 02/05/2023] Open
Abstract
Finding and constructing an osteogenic microenvironment similar to natural bone tissue has always been a frontier topic in orthopedics. We found that osteocytes are targeting cells controlling bone anabolism produced by PTH (JBMR 2017, PMID: 27704638), and osteocytes with activated Wnt signaling orchestrate bone formation and resorption (PNAS 2015, PMID: 25605937). However, methods for taking advantage of the leading role of osteocytes in bone regeneration remain unexplored. Herein, we found that the osteocytes with SKL2001-activated Wnt signaling could be an osteogenic microenvironment (SOOME) which upregulates the expression of bone transcription factor Runx2 and Bglap and promotes the differentiation of bone marrow stromal cell ST2 into osteoblasts. Interestingly, 60 μM SKL2001 treatment of osteocytic MLO-Y4 for 24 h maintained Wnt signaling activation for three days after removal, which was sufficient to induce osteoblast differentiation. Triptonide, a Wnt inhibitor, could eliminate this differentiation. Moreover, on day 5, the Wnt signaling naturally decreased to the level of the control group, indicating that this method of Wnt-signaling induction is safe to use. We quickly verified in vivo function of SOOME to a good proximation in 3D bioprinted modules composed of reciprocally printed polycaprolactone bundles (for support) and cell bundles (for bioactivity). In the cell bundles, SOOME stably supported the growth and development of ST2 cells, the 7-day survival rate was as high as 91.6%, and proliferation ability increased linearly. Similarly, SOOME greatly promoted ST2 differentiation and mineralization for 28 days. In addition, SOOME upregulated the expression of angiopoietin 1, promoted endothelial cell migration and angiogenesis, and increased node number and total length of tubes and branches. Finally, we found that the function of SOOME could be realized through the paracrine pathway. This study reveals that osteocytes with Wnt signaling activated by SKL2001 are a safe osteogenic microenvironment. Both SOOME itself and its cell-free culture supernatant can improve bioactivity for osteoblast differentiation, with composite scaffolds especially bearing application value.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiaolin Tu
- Correspondence: ; Tel.: +86-185-2382-0685
| |
Collapse
|
35
|
Xiao T, Qu H, Zeng Z, Li C, Wan J. Interleukin-35 from Interleukin-4-Stimulated Macrophages Alleviates Oxygen Glucose Deprivation/Re-oxygenation-Induced Neuronal Cell Death via the Wnt/β-Catenin Signaling Pathway. Neurotox Res 2022; 40:420-431. [PMID: 35150397 DOI: 10.1007/s12640-022-00478-7] [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: 09/22/2021] [Revised: 01/06/2022] [Accepted: 01/26/2022] [Indexed: 11/30/2022]
Abstract
Currently, brain stroke is one of the leading causes of death and disabilities. It results in depletion of oxygen and glucose in certain areas of the brain, leading to neuronal death. Re-oxygenation has been proven to attenuate neuronal damage; however, sudden oxygen supply may also cause oxidative stress and subsequent inflammation. Hence, therapies to suppress re-oxygenation-induced oxidative damage are urgently needed. Interleukin (IL)-35, an immunomodulator secreted by regulatory T cells and regulatory B cells, is proven to be a strong immune-repressive cytokine. Here, we investigated the potential role of IL-35 in a disease model of oxygen glucose deprivation/re-oxygenation (OGD/R) and found that M2 macrophage-derived IL-35 significantly alleviated inflammatory response induced by oxidative stress. Our results also showed that IL-35 treatment decreased OGD/R-induced neuronal cell death and inflammatory response. Additionally, we demonstrated that IL-35 suppresses inflammatory response via the Wnt/β-catenin signaling pathway. Hence, our findings indicate that IL-35 therapy has great potential in the treatment of OGD/R-induced oxidative damage and related inflammatory diseases.
Collapse
Affiliation(s)
- Tao Xiao
- Department of Neurosurgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Hongtao Qu
- Department of Neurosurgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhiqing Zeng
- Department of Neurosurgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Chuanghua Li
- Department of Neurosurgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Juan Wan
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, Hunan Province, China.
| |
Collapse
|
36
|
Cao C, Dai Y, Wang Z, Zhao G, Duan H, Zhu X, Wang J, Zheng M, Weng Q, Wang L, Gou W, Zhang H, Li C, Liu D, Hu Y. The role of junctional adhesion molecule-C in trophoblast differentiation and function during normal pregnancy and preeclampsia. Placenta 2022; 118:55-65. [PMID: 35032792 DOI: 10.1016/j.placenta.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/19/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Junctional adhesion molecule-C (JAM-C) is an important regulator of many physiological processes, ranging from maintenance of tight junction integrity of epithelia to regulation of cell migration, homing and proliferation. Preeclampsia (PE) is a trophoblast-related syndrome with abnormal placentation and insufficient trophoblast invasion. However, the role of JAM-C in normal pregnancy and PE pathogenesis is unknown. METHODS The expression and location of JAM-C in placentas were determined by quantitative real-time PCR (qRT-PCR), western blot and immunohistochemistry. The expression of differentiation and invasion markers were detected by qRT-PCR or western blot. The effects of JAM-C on migration and invasion of trophoblasts were examined using wound-healing and invasion assays. Additionally, a mouse model was established by injection of JAM-C-positive adenovirus to explore the effects of JAM-C in vivo. RESULTS In normal pregnancy, JAM-C was preferentially expressed on cytotrophoblast (CTB) progenitors and progressively decreased when acquiring invasion properties with gestation advance. However, in PE patients, the expression of JAM-C was upregulated in extravillous trophoblasts (EVTs) and syncytiotrophoblasts (SynTs) of placentas. It was also demonstrated that JAM-C suppressed the differentiation of CTBs into EVTs in vitro. Consistently, JAM-C inhibited the migration and invasion capacities of EVTs through GSK3β/β-catenin signaling pathway. Importantly, Ad-JAMC-infected mouse model mimicked the phenotype of human PE. DISCUSSION JAM-C plays an important role in normal placentation and upregulated JAM-C in placentas contributes to PE development.
Collapse
Affiliation(s)
- Chenrui Cao
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yimin Dai
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhiyin Wang
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Guangfeng Zhao
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Honglei Duan
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiangyu Zhu
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jingmei Wang
- Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Mingming Zheng
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qiao Weng
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Limin Wang
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Wenjing Gou
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Haili Zhang
- Department of Obstetrics and Gynecology, The First People's Hospital of Mangya, Qinghai, China
| | - Chanjuan Li
- Department of Obstetrics, Women's Hospital of Nanjing Medical University, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Heath Care Hospital, Nanjing, China
| | - Dan Liu
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| | - Yali Hu
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
37
|
Hoveizi E, Tavakol S. Differentiation of Human Wharton's Jelly Mesenchymal Stem Cells into SOX17 Expressing Cells Using a Wnt/ß-catenin Pathway Agonist on Polylactic Acid/Chitosan Nanocomposite Scaffold. CELL JOURNAL 2022; 24:55-61. [PMID: 35279960 PMCID: PMC8918270 DOI: 10.22074/cellj.2022.7622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/24/2020] [Indexed: 11/05/2022]
Abstract
Objective The β-catenin signaling pathway promises the potential for differentiation of stem cells into definitive endoderm (DE) cells as precursors of beta cells. Therefore, it can be considered as an inducer for cell replacement therapies in diabetes. The main goal of this research is to successfully culture and induce differentiation of human Wharton's jelly mesenchymal stem cells (hWJMSCs) into Sox17-expressing cells using a Wnt/β-catenin pathway agonist (SKL2001) plus nanoparticles on a polylactic acid/chitosan (PLA/Cs) nanocomposite scaffold. Materials and Methods In this experimental study, the nanocomposite was prepared through an electrospinning method and hWJMSCs were isolated through an explant technique. The morphology and the cell viability were evaluated by scanning electron microscopy (SEM) and 3-(4, 5- Dimethylthiazol-2)-2, 5-diphenyltetrazolium bromide (MTT) assay. Here, we present two differentiation protocols: the first one is induction with SKL2001; and the second one is with a combination of SKL2001 and zinc oxide nanoparticles (nZnO). Real-time quantitative reverse transcription (QRT-PCR) and immunocytochemistry analysis are carried out to examine the expression of specific markers in the differentiated cells. Results The nanocomposite had appropriate biocompatibility for cell adhesion and growth. While the hWJMSCs cultured on the PLA/Cs scaffolds differentiated into DE cells in the presence of SKL2001, introducing nZnO to their environment increased the differentiation process. Analyses of DE-specific markers including SOX17, FOXA2, and gooscoid (GSC) genes in mRNA level, indicated significantly high levels of expression in the SKL2001/nZnO group, followed by SKL2001 group compared to the control. Conclusion Our results show the beneficial effects of the Wnt/β-catenin pathway agonist in three-dimensional (3D) cultures in cell replacement therapy for diabetes.
Collapse
Affiliation(s)
- Elham Hoveizi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran,Stem Cells and Transgenic Technology Research Center (STTRC), Shahid Chamran University of Ahvaz, Ahvaz, Iran,P.O.Box: 61375Department of BiologyFaculty of ScienceShahid Chamran University of AhvazAhvazIran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Shi Z, He J, He J, Xu Y. High hydrostatic pressure (30 atm) enhances the apoptosis and inhibits the proteoglycan synthesis and extracellular matrix level of human nucleus pulposus cells via promoting the Wnt/β-catenin pathway. Bioengineered 2022; 13:3070-3081. [PMID: 35100096 PMCID: PMC8974124 DOI: 10.1080/21655979.2022.2025518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hydrostatic pressure is known to regulate bovine nucleus pulposus cell metabolism, but its mechanism in human nucleus pulposus cells (HNPCs) remains obscure, which attracts our attention and becomes the focus in this study. Specifically, HNPCs were treated with SKL2001 (an agonist in the Wnt/β-catenin pathway) or XAV-939 (an inhibitor of the Wnt/β-catenin pathway), and pressurized under the hydrostatic pressure of 1, 3 and 30 atm. The viability, apoptosis and proteoglycan synthesis of treated HNPC were assessed by CCK-8, flow cytometry and radioisotope incorporation assays. The levels of extracellular matrix, Collagen-II, matrix metalloproteinase 3 (MMP3), Wnt-3a and β-catenin were measured by toluidine blue staining, immunocytochemistry and Western blot. Appropriate hydrostatic stimulation (3 atm) enhanced the viability and proteoglycan synthesis yet inhibited the apoptosis of HNPCs, which also up-regulated extracellular matrix and Collagen-II levels, and down-regulated MMP3, Wnt-3a and β-catenin levels in treated HNPCs. Furthermore, high hydrostatic pressure (30 atm) inhibited the viability and proteoglycan synthesis, and promoted the morphological change and apoptosis of HNPCs, which also down-regulated extracellular matrix and Collagen-II levels and up-regulated MMP3, Wnt-3a and β-catenin levels. Besides, SKL2001 reversed the effects of hydrostatic pressure (3 atm) on inhibiting Wnt-3a, β-catenin, and MMP3 levels and promoting Collagen-II level in HNPC; whereas, XAV-939 reversed the effects of high hydrostatic pressure (30 atm) on promoting MMP3, Wnt-3a, and β-catenin levels and inhibiting Collagen-II level and proteoglycan synthesis of HNPCs. Collectively, high hydrostatic pressure promoted the apoptosis and inhibited the viability of HNPCs via activating the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Zongting Shi
- Department of Spine, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Jun He
- Department of Orthopedics, Zhejiang Hospital, Hangzhou City, Zhejiang Province, China
| | - Jian He
- Department of Orthopedics, Zhejiang Hospital, Hangzhou City, Zhejiang Province, China
| | - Yuan Xu
- Department of Orthopedics, Zhejiang Hospital, Hangzhou City, Zhejiang Province, China
| |
Collapse
|
39
|
Yan M, Duan X, Cai L, Zhang W, Silva MJ, Brophy RH, Rai MF. KIF26B Silencing Prevents Osseous Transdifferentiation of Progenitor/Stem Cells and Attenuates Ectopic Calcification in a Murine Model. J Bone Miner Res 2022; 37:349-368. [PMID: 34787331 PMCID: PMC11684001 DOI: 10.1002/jbmr.4473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 01/28/2023]
Abstract
Ectopic calcification is an osteogenic process that leads to the formation of inappropriate bone within intra-articular soft tissues, often in response to injury or surgery. The molecular mechanisms governing this phenotype have yet to be determined. Using a population genetics approach, we identified an association of the kinesin superfamily member 26b (Kif26b) with injury-induced ectopic calcification through quantitative trait locus analysis of recombinant inbred mouse strains, consistent with a genomewide association study that identified KIF26B as a severity locus for ectopic calcification in patients with hip osteoarthritis. Despite these associations of KIF26B with ectopic calcification, its mechanistic role and functional implications have not yet been fully elucidated. Here, we aim to decipher the functional role of KIF26B in osseous and chondrogenic transdifferentiation of human and murine progenitor/stem cells and in a murine model of non-invasive injury-induced intra-articular ectopic calcification. We found that KIF26B ablation via lentivirus-mediated shRNA significantly arrested osteogenesis of progenitor/stem cells and suppressed the expression of typical osteogenic marker genes. Conversely, KIF26B loss-of-function increased chondrogenesis as demonstrated by enhanced Safranin-O staining and by the elevated expression of chondrogenic marker genes. Furthermore, cell function analysis revealed that KIF26B knockdown significantly decreased cell viability and proliferation and induced cellular apoptosis. Mechanistically, loss of osteogenesis was reverted by the addition of a Wnt agonist, SKL2001, demonstrating a role of KIF26B in canonical Wnt/β-catenin signaling. Finally, intra-articular delivery of Kif26b shRNA in B6-129SF2/J mice significantly hampered the development of intra-articular ectopic calcification at 8 weeks after injury compared with mice treated with non-target scrambled shRNA. In summary, these observations highlight that KIF26B plays a crucial role in ectopic bone formation by repressing osteogenesis, but not chondrogenesis, potentially via modulating Wnt/β-catenin signaling. These findings establish KIF26B as a critical determinant of the osteogenic process in pathologic endochondral bone formation and an actionable target for pharmacotherapy to mitigate ectopic calcification (and heterotopic ossification). © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Mingming Yan
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University, School of Medicine, St. Louis, MO, United States
- Department of Orthopedic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Duan
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University, School of Medicine, St. Louis, MO, United States
| | - Lei Cai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University, School of Medicine, St. Louis, MO, United States
| | - Weili Zhang
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University, School of Medicine, St. Louis, MO, United States
| | - Matthew J. Silva
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University, School of Medicine, St. Louis, MO, United States
| | - Robert H. Brophy
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University, School of Medicine, St. Louis, MO, United States
| | - Muhammad Farooq Rai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University, School of Medicine, St. Louis, MO, United States
- Department of Cell Biology and Physiology, Washington University, School of Medicine, St. Louis, MO, United States
| |
Collapse
|
40
|
Sung BY, Lin YH, Kong Q, Shah PD, Glick Bieler J, Palmer S, Weinhold KJ, Chang HR, Huang H, Avery RK, Schneck J, Chiu YL. Wnt activation promotes memory T cell polyfunctionality via epigenetic regulator PRMT1. J Clin Invest 2022; 132:e140508. [PMID: 35040433 PMCID: PMC8759796 DOI: 10.1172/jci140508] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/21/2021] [Indexed: 12/11/2022] Open
Abstract
T cell polyfunctionality is a hallmark of protective immunity against pathogens and cancer, yet the molecular mechanism governing it remains mostly elusive. We found that canonical Wnt agonists inhibited human memory CD8+ T cell differentiation while simultaneously promoting the generation of highly polyfunctional cells. Downstream effects of Wnt activation persisted after removal of the drug, and T cells remained polyfunctional following subsequent cell division, indicating the effect is epigenetically regulated. Wnt activation induced a gene expression pattern that is enriched with stem cell-specific gene signatures and upregulation of protein arginine methyltransferase 1 (PRMT1), a known epigenetic regulator. PRMT1+CD8+ T cells are associated with enhanced polyfunctionality, especially the ability to produce IL-2. In contrast, inhibition of PRMT1 ameliorated the effects of Wnt on polyfunctionality. Chromatin immunoprecipitation revealed that H4R3me2a, a permissive transcription marker mediated by PRMT1, increased at the IL-2 promoter loci following Wnt activation. In vivo, Wnt-treated T cells exhibited superior polyfunctionality and persistence. When applied to cytomegalovirus (CMV) donor-seropositive, recipient-seronegative patients (D+/R-) lung transplant patient samples, Wnt activation enhanced CMV-specific T cell polyfunctionality, which is important in controlling CMV diseases. These findings reveal a molecular mechanism governing T cell polyfunctionality and identify PRMT1 as a potential target for T cell immunotherapy.
Collapse
Affiliation(s)
- Bo-Yi Sung
- Institute of Cell Engineering and
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Microbiology and Immunology
- Department of Biomedical Engineering, and
| | - Yi-Hsin Lin
- Department of Biomedical Engineering, and
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | | | - Pali D. Shah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Joan Glick Bieler
- Institute of Cell Engineering and
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Kent J. Weinhold
- Department of Surgery, and Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robin K. Avery
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jonathan Schneck
- Institute of Cell Engineering and
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Medicine and Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland. USA
| | - Yen-Ling Chiu
- Institute of Cell Engineering and
- Graduate Institute of Medicine and Graduate Program in Biomedical Informatics, Yuan Ze University, Taoyuan, Taiwan
- Department of Medical Research, Far Eastern Memorial Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
41
|
Cheng X, Qin L, Deng L, Zhu X, Li Y, Wu X, Zheng Y. SNX-2112 Induces Apoptosis and Inhibits Proliferation, Invasion, and Migration of Non-Small Cell Lung Cancer by Downregulating Epithelial-Mesenchymal Transition via the Wnt/β-Catenin Signaling Pathway. J Cancer 2021; 12:5825-5837. [PMID: 34475996 PMCID: PMC8408115 DOI: 10.7150/jca.56640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 06/20/2021] [Indexed: 12/30/2022] Open
Abstract
Lung cancer is the most frequent malignant tumor, and non-small cell lung cancer (NSCLC) is responsible for substantial mortality worldwide. The small molecule SNX-2112 was recently shown to critically effect the proliferation and apoptosis of tumor cells. Nevertheless, the precise mechanism by which SNX-2112 affects NSCLC remains poorly understood. Therefore, we investigated the function of SNX-2112 in NSCLC. We verified that SNX-2112 promoted apoptosis and suppressed the proliferation, invasion, and migration of A549 and H520 NSCLC cells in vitro. We further verified the potential mechanism of SNX-2112 in NSCLC. The changes in the protein levels demonstrated that SNX-2112 inhibited the epithelial-mesenchymal transition (EMT) (increased E-cadherin and decreased N-cadherin and vimentin) and the Wnt/β-catenin signaling pathway (glycogen synthase kinase (GSK) 3β and phosphorylated (p)-β-catenin increased, β-catenin and p-GSK3β decreased) in NSCLC cells. These results were verified by rescue experiments using a Wnt/β-catenin pathway agonist. We also established a tumor xenograft model and confirmed that SNX-2112 reduced tumor growth and proliferation and enhanced necrosis and apoptosis in a NSCLC model in vivo. In conclusion, the current study is the first to discover the mechanism of SNX-2112 in NSCLC. SNX-2112 induced apoptosis and also inhibited the proliferation, invasion, and migration of NSCLC cells by downregulating EMT via the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xiaozhen Cheng
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, No.78 Heng-Zhi-Gang Road, Yue Xiu District, Guangzhou 510095, China.,Department of Oncology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Hainan Province, 570208, China
| | - Lingyu Qin
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, No.78 Heng-Zhi-Gang Road, Yue Xiu District, Guangzhou 510095, China.,Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253 Industry Road, Guangzhou 510282, China
| | - Lian Deng
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, No.78 Heng-Zhi-Gang Road, Yue Xiu District, Guangzhou 510095, China.,Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253 Industry Road, Guangzhou 510282, China
| | - Xiongjie Zhu
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, No.78 Heng-Zhi-Gang Road, Yue Xiu District, Guangzhou 510095, China.,Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253 Industry Road, Guangzhou 510282, China
| | - Ying Li
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, No.78 Heng-Zhi-Gang Road, Yue Xiu District, Guangzhou 510095, China.,Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253 Industry Road, Guangzhou 510282, China
| | - Xiaoran Wu
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, No.78 Heng-Zhi-Gang Road, Yue Xiu District, Guangzhou 510095, China.,Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253 Industry Road, Guangzhou 510282, China
| | - Yanfang Zheng
- Medical Oncology Department, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, No.78 Heng-Zhi-Gang Road, Yue Xiu District, Guangzhou 510095, China.,Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253 Industry Road, Guangzhou 510282, China
| |
Collapse
|
42
|
Bonnet C, Brahmbhatt A, Deng SX, Zheng JJ. Wnt signaling activation: targets and therapeutic opportunities for stem cell therapy and regenerative medicine. RSC Chem Biol 2021; 2:1144-1157. [PMID: 34458828 PMCID: PMC8341040 DOI: 10.1039/d1cb00063b] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022] Open
Abstract
Wnt proteins are secreted morphogens that play critical roles in embryonic development, stem cell proliferation, self-renewal, tissue regeneration and remodeling in adults. While aberrant Wnt signaling contributes to diseases such as cancer, activation of Wnt/β-catenin signaling is a target of interest in stem cell therapy and regenerative medicine. Recent high throughput screenings from chemical and biological libraries, combined with improved gene expression reporter assays of Wnt/β-catenin activation together with rational drug design, led to the development of a myriad of Wnt activators, with different mechanisms of actions. Among them, Wnt mimics, antibodies targeting Wnt inhibitors, glycogen-synthase-3β inhibitors, and indirubins and other natural product derivatives are emerging modalities to treat bone, neurodegenerative, eye, and metabolic disorders, as well as prevent ageing. Nevertheless, the creation of Wnt-based therapies has been hampered by challenges in developing potent and selective Wnt activators without off-target effects, such as oncogenesis. On the other hand, to avoid these risks, their use to promote ex vivo expansion during tissue engineering is a promising application.
Collapse
Affiliation(s)
- Clémence Bonnet
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- INSERM, UMRS1138, Team 17, From Physiopathology of Ocular Diseases to Clinical Development, Paris University, Centre de Recherche des Cordeliers, and Cornea Departement, Cochin Hospital, AP-HP F-75014 Paris France
| | - Anvi Brahmbhatt
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
| | - Sophie X Deng
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- Molecular Biology Institute, University of California Los Angeles CA USA
| | - Jie J Zheng
- Stein Eye Institute, University of California Los Angeles CA USA +1-3107947906 +1-3102062173
- Molecular Biology Institute, University of California Los Angeles CA USA
| |
Collapse
|
43
|
Wang T, Zhang T, Tang Y, Wang H, Wei Q, Lu Y, Yao J, Qu Y, Cao X. Oxysterol-binding protein-like 2 contributes to the developmental progression of preadipocytes by binding to β-catenin. Cell Death Discov 2021; 7:109. [PMID: 34001864 PMCID: PMC8129138 DOI: 10.1038/s41420-021-00503-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Oxysterol-binding protein-like 2 (OSBPL2), also known as oxysterol-binding protein-related protein (ORP) 2, is a member of lipid transfer protein well-known for its role in regulating cholesterol homeostasis. A recent study reported that OSBPL2/ORP2 localizes to lipid droplets (LDs) and is associated with energy metabolism and obesity. However, the function of OSBPL2/ORP2 in adipocyte differentiation is poorly understood. Here, we report that OSBPL2/ORP2 contributes to the developmental progression of preadipocytes. We found that OSBPL2/ORP2 binds to β-catenin, a key effector in the Wnt signaling pathway that inhibits adipogenesis. This complex plays a role in regulating the protein level of β-catenin only in preadipocytes, not in mature adipocytes. Our data further indicated that OSBPL2/ORP2 mediates the transport of β-catenin into the nucleus and thus regulates target genes related to adipocyte differentiation. Deletion of OSBPL2/ORP2 markedly reduces β-catenin both in the cytoplasm and in the nucleus, promotes preadipocytes maturation, and ultimately leads to obesity-related characteristics. Altogether, we provide novel insight into the function of OSBPL2/ORP2 in the developmental progression of preadipocytes and suggest OSBPL2/ORP2 may be a potential therapeutic target for the treatment of obesity-related diseases.
Collapse
Affiliation(s)
- Tianming Wang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Tianyu Zhang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Youzhi Tang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Hongshun Wang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Qinjun Wei
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Yajie Lu
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Jun Yao
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Yuan Qu
- Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing, China
| | - Xin Cao
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China. .,Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
44
|
Acute myeloid leukemia-induced remodeling of the human bone marrow niche predicts clinical outcome. Blood Adv 2021; 4:5257-5268. [PMID: 33108453 DOI: 10.1182/bloodadvances.2020001808] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022] Open
Abstract
Murine models of myeloid neoplasia show how leukemia infiltration alters the hematopoietic stem cell (HSC) niche to reinforce malignancy at the expense of healthy hematopoiesis. However, little is known about the bone marrow architecture in humans and its impact on clinical outcome. Here, we dissect the bone marrow niche in patients with acute myeloid leukemia (AML) at first diagnosis. We combined immunohistochemical stainings with global gene expression analyses from these AML patients and correlated them with clinical features. Mesenchymal stem and progenitor cells (MSPCs) lost quiescence and significantly expanded in the bone marrow of AML patients. Strikingly, their HSC- and niche-regulating capacities were impaired with significant inhibition of osteogenesis and bone formation in a cell contact-dependent manner through inhibition of cytoplasmic β-catenin. Assessment of bone metabolism by quantifying peripheral blood osteocalcin levels revealed 30% lower expression in AML patients at first diagnosis than in non-leukemic donors. Furthermore, patients with osteocalcin levels ≤11 ng/mL showed inferior overall survival with a 1-year survival rate of 38.7% whereas patients with higher osteocalcin levels reached a survival rate of 66.8%. These novel insights into the human AML bone marrow microenvironment help translate findings from preclinical models and detect new targets which might pave the way for niche-targeted therapies in AML patients.
Collapse
|
45
|
Upregulation of lnc-ZNF281 Inhibits the Progression of Glioma via the AKT/GSK-3 β/ β-Catenin Signaling Pathway. J Immunol Res 2021; 2021:5573071. [PMID: 34056009 PMCID: PMC8131163 DOI: 10.1155/2021/5573071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/14/2021] [Accepted: 04/26/2021] [Indexed: 11/17/2022] Open
Abstract
The purpose of this study is to elucidate the roles and potential underlying mechanisms of long noncoding RNA lnc-ZNF281 in glioma. We performed qRT-PCR to detect the expression levels of lnc-ZNF281 in glioma tissues. The effects of lnc-ZNF281 on the proliferative and migrative abilities of T98G and HS683 glioma cells were examined by cell proliferation assay, colony formation assay, wound-healing assay, and transwell assay. Also, the effects of lnc-ZNF281 on AKT/GSK-3β/β-catenin pathway were analyzed. The results showed that the expression of lnc-ZNF281 in glioma tissues was decreased compared with normal tissues. lnc-ZNF281 overexpression inhibited the proliferative and migrative abilities of glioma cells, while lnc-ZNF281 knockdown obtained the opposite findings. Besides, overexpression of lnc-ZNF281 in glioma cells inactivated the AKT/GSK-3β/β-catenin signaling pathway. Furthermore, β-catenin activation reversed the suppressive effects of lnc-ZNF281 on glioma cells. Taken together, lnc-ZNF281 inhibits glioma cell proliferation and migration via AKT/GSK-3β/β-catenin pathway and may serve as a potential target for glioma treatment.
Collapse
|
46
|
Tang N, Xu S, Song T, Qiu Y, He J, Fu X. Zinc finger protein 91 accelerates tumour progression by activating β-catenin signalling in pancreatic cancer. Cell Prolif 2021; 54:e13031. [PMID: 33755268 PMCID: PMC8088462 DOI: 10.1111/cpr.13031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES ZFP91, an E3 ligase, has been reported to possess cancer-promoting functions. This study aimed to elucidate the exact role of ZFP91 in tumour progression of pancreatic cancer and underlying mechanisms. MATERIALS AND METHODS We analysed the correlation between ZFP91 expression and pancreatic cancer through TCGA and GEO data sets. Growth curve, colony formation, wound healing and transwell invasion assays were conducted to evaluate proliferation, migration and invasion of lentivirus transfected pancreatic cancer cells. GSEA and Western blot analysis were performed to validate the regulatory effect of ZFP91 on β-catenin. Drug response curve and orthotopic implantation model reflected the sensitivity of chemotherapies. RESULTS ZFP91 overexpression is prevalent in pancreatic cancer and negatively correlated with overall survival. ZFP91 knock-down attenuated proliferation, migration and invasion of pancreatic cancer cells. β-catenin was a downstream gene of ZFP91, and its agonist could reverse the phenotype. ZFP91 promoted EMT and chemoresistance in pancreatic cancer. CONCLUSIONS We demonstrated that ZFP91 promoted pancreatic cancer proliferation, migration and invasion through activating β-catenin signalling. EMT and chemoresistance were also regulated by ZFP91. ZFP91 might be a potential therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Neng Tang
- Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Shanshan Xu
- Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Taiyu Song
- Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Yudong Qiu
- Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Jian He
- Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Xiao Fu
- Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| |
Collapse
|
47
|
Lnc GNG12-AS1 knockdown suppresses glioma progression through the AKT/GSK-3β/β-catenin pathway. Biosci Rep 2021; 40:225952. [PMID: 32735016 PMCID: PMC7435023 DOI: 10.1042/bsr20201578] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/08/2020] [Accepted: 07/29/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are increasingly being regarded as regulators of glioma development. Notably, some studies report that GNG12-AS1 plays important functions and molecular mechanism in breast cancer, but there are no existing studies in glioma. OBJECTIVE To analyze the biological functions and potential mechanisms of GNG12-AS1 in glioma. METHODS We detected the expression of GNG12-AS1 in glioma tissues through analyzing TCGA data as well as our clinical samples. We then evaluated cell proliferation through MTT assay and colony formation and cell migration by transwell assay, wound healing assay and single cell tracking assay. After, we analyzed the effects of the AKT/GSK-3β/β-catenin through Western blotting and utilized the β-catenin agonist SKL2001 for the rescue experiment. RESULTS GNG12-AS1 was highly expressed in glioma tissues. The silence of GNG12-AS1 inhibited the proliferation, migration and epithelial-mesenchymal transition of glioma cells, and reduced the activity of the AKT/GSK-3β/β-catenin pathway. Notably, SKL2001 could reverse cell migration as well as β-catenin expression in glioma cells with lower GNG12-AS1 expression. CONCLUSIONS GNG12-AS1 regulates proliferation and migration of glioma cells through the AKT/GSK-3β/β-catenin signaling and can perhaps be a new target for the treatment of glioma.
Collapse
|
48
|
Wang Z, Li Z, Ji H. Direct targeting of β-catenin in the Wnt signaling pathway: Current progress and perspectives. Med Res Rev 2021; 41:2109-2129. [PMID: 33475177 DOI: 10.1002/med.21787] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/30/2020] [Accepted: 01/05/2021] [Indexed: 12/28/2022]
Abstract
Aberrant activation of the Wnt/β-catenin signaling circuit is associated with cancer recurrence and relapse, cancer invasion and metastasis, and cancer immune evasion. Direct targeting of β-catenin, the central hub in this signaling pathway, is a promising strategy to suppress the hyperactive β-catenin signaling but has proven to be highly challenging. Substantial efforts have been made to discover compounds that bind with β-catenin, block β-catenin-mediated protein-protein interactions, and suppress β-catenin signaling. Herein, we characterize potential small-molecule binding sites in β-catenin, summarize bioactive small molecules that directly target β-catenin, and review structure-based inhibitor optimization, structure-activity relationship, and biological activities of reported inhibitors. This knowledge will benefit future inhibitor development and β-catenin-related drug discovery.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Zilu Li
- Department of Chemistry, University of South Florida, Tampa, Florida, USA
| | - Haitao Ji
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Department of Chemistry, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
49
|
Progress and Challenges of Amniotic Fluid Derived Stem Cells in Therapy of Ischemic Heart Disease. Int J Mol Sci 2020; 22:ijms22010102. [PMID: 33374215 PMCID: PMC7794729 DOI: 10.3390/ijms22010102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/13/2020] [Accepted: 12/18/2020] [Indexed: 02/08/2023] Open
Abstract
Cardiovascular disease is the leading cause of deaths worldwide, claiming an estimated total of 17.9 million lives each year, of which one-third of the people are under the age of 70 years. Since adult cardiomyocytes fail to regenerate, the heart loses the ability to repair itself after an injury, making patients with heart disease suffer from poor prognosis. Pluripotent stem cells have the ability to differentiate into cardiomyocytes in vitro through a well-established process, which is a new advancement in cardiac regeneration therapy. However, pluripotent stem cell-derived cardiomyocytes have certain drawbacks, such as the risk of arrhythmia and immune incompatibility. Thus, amniotic fluid stem cells (AFSCs), a relatively novel source of stem cells, have been exploited for their ability of pluripotent differentiation. In addition, since AFSCs are weakly positive for the major histocompatibility class II molecules, they may have high immune tolerance. In summary, the possibility of development of cardiomyocytes from AFSCs, as well as their transplantation in host cells to produce mechanical contraction, has been discussed. Thus, this review article highlights the progress of AFSC therapy and its application in the treatment of heart diseases in recent years.
Collapse
|
50
|
Costa R, Muccioli S, Brillo V, Bachmann M, Szabò I, Leanza L. Mitochondrial dysfunction interferes with neural crest specification through the FoxD3 transcription factor. Pharmacol Res 2020; 164:105385. [PMID: 33348025 DOI: 10.1016/j.phrs.2020.105385] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 11/28/2022]
Abstract
The neural crest is an important group of cells with pluripotency and migratory ability that is crucially involved in tissue and cell specification during development. Craniofacial shaping, sensory neurons, body asymmetry, and pigmentation are linked to neural crest functionality. Despite its prominent role in embryogenesis, neural crest specification as well as the possible part mitochondria play in such a process remains unclarified. Mitochondria are important organelles not only for respiration, but also for regulation of cell proliferation, differentiation and death. Modulation of mitochondrial fitness and depletion of mitochondrial ATP synthesis has been shown to down-regulate Wnt signaling, both in vitro and in vivo. Since Wnt signaling is one of the crucial players during neural crest induction/specification, we hypothesized a signaling cascade connecting mitochondria to embryonic development and neural crest migration and differentiation. Here, by using pharmacological and genetic modulators of mitochondrial function, we provide evidence that a crosstalk between mitochondrial energy homeostasis and Wnt signaling is important in the development of neural crest-derived tissues. Furthermore, our results highlight the possibility to modulate neural crest cell specification by tuning mitochondrial metabolism via FoxD3, an important transcription factor that is regulated by Wnt. FoxD3 ensures the correct embryonic development and contributes to the maintenance of cell stemness and to the induction of epithelial-to-mesenchymal transition. In summary, our work offers new insights into the molecular mechanism of action of FoxD3 and demonstrates that mitochondrial fitness is linked to the regulation of this important transcription factor via Wnt signaling in the context of neural crest specification.
Collapse
Affiliation(s)
- Roberto Costa
- Department of Biology, University of Padova, Padova, Italy
| | | | | | | | - Ildikò Szabò
- Department of Biology, University of Padova, Padova, Italy
| | - Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy.
| |
Collapse
|