1
|
Chang YH, Lee YC, Chen SH, Fang SY, Cheng TP, Chi CH, Tsai KC, Chen PJ, Hung HY. Discovery of a novel C2-functionalized chromen-4-one scaffold for the development of p38α MAPK signaling inhibitors to mitigate neutrophilic inflammatory responses. Biochem Pharmacol 2025; 235:116806. [PMID: 39956209 DOI: 10.1016/j.bcp.2025.116806] [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: 10/08/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
Neutrophil dysregulation is implicated in a spectrum of inflammatory pathologies, suggesting the potential for targeting neutrophilic hyperactivation as a pharmacological strategy to manage inflammatory disorders. Building upon prior research where 2-thiolphenoxychromone derivatives were found to inhibit neutrophilic generation of superoxide anions, this study focused on exploring the structure-activity relationship (SAR) of different C2 bridging moieties and anti-inflammatory effects using bioisosteric replacements and scaffold-hopping approaches. Among various chemotypes, the N-(4-oxo-4H-chromen-2-yl)benzenesulfonamide derivatives emerged as robust inhibitors of both superoxide anion generation and elastase release from fMLF-activated human neutrophils, with IC50 values in the single-digit micromolar range. Leveraging a forward pharmacology approach through computational prediction, compound 15b, a representative within this active molecular class, was discovered to exert these anti-inflammatory functions by blocking the p38α mitogen-activated protein kinase (MAPK) signaling cascade. This responded to a significant reduction in p38α MAPK and its downstream MK2 phosphorylation in activated neutrophils treated with 15b, with no apparent impact on extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and protein kinase B (AKT) phosphorylation levels. Additionally, this molecule exhibited inhibitory potential on intracellular reactive oxygen species (ROS) production, granule exocytosis, and chemotactic responses. Collectively, this study provides a novel skeleton for the development of inhibitors targeting the p38α MAPK pathway to mitigate neutrophilic inflammation.
Collapse
Affiliation(s)
- Yi-Han Chang
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Yi-Chen Lee
- Department of Nutrition Therapy, E-Da Cancer Hospital, I-Shou University, Kaohsiung 824, Taiwan; Department of Nutrition Therapy, E-Da Hospital, I-Shou University, Kaohsiung 824, Taiwan; Department of Nutrition Therapy, E-Da Dachang Hospital, I-Shou University, Kaohsiung 824, Taiwan; Department of Nutrition, College of Medicine, I-Shou University, Kaohsiung 824, Taiwan
| | - Shun-Hua Chen
- School of Nursing, Fooyin University, Kaohsiung 831, Taiwan
| | - Shu-Yen Fang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Tzu-Peng Cheng
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ching-Ho Chi
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112, Taiwan
| | - Keng-Chang Tsai
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112, Taiwan
| | - Po-Jen Chen
- Department of Medical Research, E-DA Hospital, I-Shou University, Kaohsiung 824, Taiwan; Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Hsin-Yi Hung
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
2
|
Liu X, Yu D, Yu Z, Su S, Jiang M, Zhao C. LHPP-P38 MAPK/ERK-ETS1 Axis Negative Feedback Signaling Restrains Progression in Breast Cancer. Cancer Sci 2025; 116:923-935. [PMID: 39789996 PMCID: PMC11967269 DOI: 10.1111/cas.16448] [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: 08/23/2024] [Revised: 12/03/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
Invasion and metastasis are major causes of mortality in breast cancer (BRCA) patients. LHPP, known for its tumor-suppressive effects, has an undefined role in BRCA. We found reduced LHPP protein in BRCA tissues, with lower levels correlating with poor patient outcomes. In vitro studies show LHPP inhibits BRCA cell proliferation, migration, invasion, and stemness. In vivo xenograft models support LHPP's role in curbing tumorigenesis and lung metastasis. Mechanistically, LHPP interacts with ERK and P38 MAPK, leading to their dephosphorylation and suppression of the MAPK pathway. We also reveal ETS1, a MAPK effector, repressing LHPP mRNA transcription, suggesting a LHPP-P38 MAPK/ERK-ETS1 negative feedback loop as a key regulatory mechanism in controlling BRCA invasion and metastasis.
Collapse
Affiliation(s)
- Xu Liu
- Abdominal Radiotherapy Department, Harbin Medical University Cancer HospitalHarbin Medical UniversityHarbinHeilongjiang ProvinceChina
| | - Deyang Yu
- Radiophysics Department, Harbin Medical University Cancer HospitalHarbin Medical UniversityHarbinHeilongjiang ProvinceChina
| | - Zhen Yu
- Abdominal Radiotherapy Department, Harbin Medical University Cancer HospitalHarbin Medical UniversityHarbinHeilongjiang ProvinceChina
| | - Sisi Su
- Abdominal Radiotherapy Department, Harbin Medical University Cancer HospitalHarbin Medical UniversityHarbinHeilongjiang ProvinceChina
| | - Meixia Jiang
- Abdominal Radiotherapy Department, Harbin Medical University Cancer HospitalHarbin Medical UniversityHarbinHeilongjiang ProvinceChina
| | - Chunbo Zhao
- Abdominal Radiotherapy Department, Harbin Medical University Cancer HospitalHarbin Medical UniversityHarbinHeilongjiang ProvinceChina
| |
Collapse
|
3
|
Li H, Zhang Y, Peh HY. Interferon regulatory factor 3 beyond innate immunity: Regulation in obesity and metabolic disorders. Semin Immunol 2025; 78:101948. [PMID: 40156960 DOI: 10.1016/j.smim.2025.101948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Interferon regulatory factor 3 (IRF3) is a transcription factor known primarily for its role in antiviral immunity via regulation of type I interferons (IFNs). Recent research has broadened its significance to encompass metabolic disorders, particularly obesity and diabetes. Obesity is characterized by chronic low-grade inflammation, insulin resistance, and metabolic dysfunction, all of which are increasingly found to be associated with immune signaling pathways. IRF3 has emerged as an important regulator in the development of obesity and type 2 diabetes (T2D), predominantly through its regulation of inflammatory cytokines production in various cells in adipose tissue. In obese individuals, IRF3 is activated in the adipocytes and adipose tissue macrophages, to promote the expression of inflammatory cytokines, thereby contributing to chronic inflammation and exacerbating insulin resistance. Moreover, IRF3 has been linked to mitochondrial dysfunction in hepatic disorders, further amplifying metabolic stress and imbalances associated with obesity. The growing evidence suggests that IRF3 is an important mediator in both immune and metabolic pathways, highlighting its potential as a target for the development of therapeutic interventions for obesity-related inflammation and metabolic dysfunction.
Collapse
Affiliation(s)
- Heng Li
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore; NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore; NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore.
| | - Hong Yong Peh
- NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore; Singapore Lipidomics Incubator, Life Science Institute, National University of Singapore, Singapore 117456, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| |
Collapse
|
4
|
Xiong Y, Zhang X, Xie W, Yin Y, Qian Y, Ying X, Zheng X, Wang X. DUSP4 inhibited tumor cell proliferation by downregulating glycolysis via p-ERK/p-PGK1 signaling in ovarian cancer. Cancer Cell Int 2025; 25:87. [PMID: 40082940 PMCID: PMC11908039 DOI: 10.1186/s12935-025-03722-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 03/01/2025] [Indexed: 03/16/2025] Open
Abstract
Ovarian cancer (OC) remains a leading cause of gynecological cancer-related mortality, with poor prognosis and limited therapeutic options, underscoring the urgent need for a deeper understanding of OC biology. In this study, we identified a marked reduction in dual-specificity phosphatase 4 (DUSP4) expression in OC tissues compared to benign ovarian masses, with even further decreases observed in metastatic lesions. Moreover, DUSP4 expression varied among OC subtypes, with the lowest levels observed in serous ovarian cancer, and was associated with P53 and KI67 protein levels, altered TP53 mutation rates, advanced tumor stages, and poorer prognosis. Functional experiments demonstrated that DUSP4 overexpression suppressed OC cell proliferation, migration, and invasion in vitro. Phosphoproteomic profiling via LC-MS/MS analysis identified the MAPK pathway and cellular metabolism as key downstream targets of DUSP4. Notably, DUSP4 overexpression reduced phosphorylation of PGK1 at Ser203, a critical regulator of anaerobic glycolysis, and decreased its mitochondrial localization, leading to reduced lactate production and increased ROS levels. Mechanistically, DUSP4 dephosphorylated p-ERK, disrupting its interaction with PGK1 and subsequently reducing PGK1 S203 phosphorylation. In vivo, DUSP4 overexpression significantly inhibited tumor growth in mouse models, accompanied by decreased p-ERK and PGK1 S203 levels. These findings highlight a regulatory axis involving DUSP4, p-ERK, and PGK1, through which DUSP4 modulates glycolysis and tumor progression. This study establishes DUSP4 as a prognostic biomarker and a potential therapeutic target for OC, offering new insights into its role in tumor metabolism and growth.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Xiaoqian Zhang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Weiwei Xie
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Yujia Yin
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Yujing Qian
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Xiang Ying
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Xiaocui Zheng
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
5
|
Shikata S, Kikkawa K, Fujimuro M, Sekine Y. Dual-specific phosphatase DUSP21 is a novel negative feedback regulator for STAT3. Biochem Biophys Res Commun 2025; 752:151488. [PMID: 39961235 DOI: 10.1016/j.bbrc.2025.151488] [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: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/24/2025]
Abstract
Dual-specificity phosphatases (DUSPs) catalyze the dephosphorylation of tyrosine and serine/threonine residues in target proteins. Atypical DUSPs (aDUSPs) lack substrate-binding motifs, suggesting their potential to target a diverse array of substrates. This study demonstrated that DUSP21, an aDUSP, is induced by leukemia inhibitory factor (LIF) in HeLa cells and acts as a negative regulator of LIF-induced signal transducer and activator of transcription 3 (STAT3) activation. Overexpressed DUSP21 co-localized and interacted with STAT3 in HeLa cells. Recombinant DUSP21 directly dephosphorylated STAT3 in vitro. Additionally, DUSP21 overexpression modulated STAT3-dependent growth of Ba/F3-G133 cells. These findings indicate that LIF-induced DUSP21 exerts an inhibitory effect on the LIF/STAT3 signaling pathway, thereby functioning as a suppressor of STAT3-mediated transcriptional activity.
Collapse
Affiliation(s)
- Shota Shikata
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, 607-8412, Japan
| | - Kazuna Kikkawa
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, 607-8412, Japan
| | - Masahiro Fujimuro
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, 607-8412, Japan
| | - Yuichi Sekine
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, 607-8412, Japan.
| |
Collapse
|
6
|
Wang S, Peng X, Zhu Q, Lu S, Hu P, Kim IH, Liu HY, Ennab W, Muniyappan M, Cai D. Lithocholic acid attenuates DON-induced inflammatory responses via epigenetic regulation of DUSP5 and TRAF5 in porcine intestinal epithelial cells. Front Vet Sci 2025; 12:1493496. [PMID: 40093618 PMCID: PMC11906417 DOI: 10.3389/fvets.2025.1493496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Deoxynivalenol (DON) is the most common mycotoxin that frequently contaminates human food and animal feed, resulting in intestinal diseases and systemic immunosuppression. Lithocholic acid (LCA) exhibits various pharmacological activities. RNA-seq and ChIP-qPCR analysis were used in the current study to investigate the protective mechanism of LCA for DON-induced inflammatory Responses via Epigenetic Regulation of DUSP5 and TRAF5 in porcine ileal epithelial cell lines (IPI-2I) cells. The IPI-2I cells were treated with the vehicle group, 250 ng/mL DON, 20 μmol/L LCA, 250 ng/mL DON+ 20 μmol/L LCA for 24 h could induce inflammatory Responses via Epigenetic Regulation of DUSP5 and TRAF5 in IPI-2I cells. By analyzing the transcriptional profiles of DON and LCA-treated IPI-2I, we observed significant transcriptional changes in IPI-2I cells. Further analysis of up-and down-regulated differential genes revealed the enrichment of pathways closely related to inflammation and apoptosis, such as the MAPK signaling pathway, IL17 signaling pathway, and Wnt signaling pathway. An upregulated (p < 0.05) relative mRNA expression level of RAP1B, GDNF, FGF2, IL1R1, RAPGEF2, DUSP5, TGFB3, CACNA1G, TEK and RPS6KA2 were noted in IPI-2I exposed to DON. DON-exposed IPI-2I cells dramatically enhanced (p < 0.05) histone marks associated with transcriptional activation, H3K9ac, H3K18ac, H3K27ac, H3K4me1, H3K9bhb, H3K18bhb Pol-II and Ser5 Pol-II at the enhancers of DUSP5 and TRAF5. Overall, our findings provide a theoretical basis for understanding the mechanism of action of LCA in attenuating DON-induced intestinal injury and for better understanding the potential of LCA as a treatment or prevention of mycotoxin-associated intestinal diseases in swine production.
Collapse
Affiliation(s)
- Shiqi Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xiaoxu Peng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Qi Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Sichen Lu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ping Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - In Ho Kim
- Department of Animal Resource and Science, Dankook University, Cheonan, Choongnam, Republic of Korea
| | - Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Wael Ennab
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Madesh Muniyappan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
7
|
Zhu L, Dong Y, Guo H, Qiu J, Guo J, Hu Y, Pan C. Murine Model Insights: Identifying Dusp15 as a Novel Biomarker for Diabetic Cardiomyopathy Uncovered Through Integrated Omics Analysis and Experimental Validation. Diabetes Metab Syndr Obes 2025; 18:515-527. [PMID: 39990179 PMCID: PMC11847420 DOI: 10.2147/dmso.s501563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/08/2025] [Indexed: 02/25/2025] Open
Abstract
Background Diabetic Cardiomyopathy (DCM) is a heart condition that arises specifically from diabetes mellitus, characterized by cardiac dysfunction in the absence of coronary artery disease or hypertension. The prevalence of DCM is rising in tandem with the global increase in diabetes, necessitating the development of early diagnostic markers and therapeutic targets. This study integrates bioinformatics analysis with experimental validation to identify potential biomarkers for DCM. Methods We performed gene expression data mining from the Gene Expression Omnibus (GEO) database. We employed Weighted Gene Co-expression Network Analysis (WGCNA) coupled with machine learning techniques to sift through hub differentially expressed genes (DEGs). Functional enrichment and protein-protein interaction (PPI) network analysis were also conducted to pinpoint key genes functions. Subsequent in vitro and in vivo experiments were performed to validate the findings. Results Our analysis revealed six core genes significantly associated with DCM. The expression of Dusp15 was notably downregulated and validated in both high-glucose cultured cardiomyocytes and DCM animal models, suggesting its potential role in DCM pathogenesis. Conclusion The integration of bioinformatics with experimental approaches has identified Dusp15 as a promising candidate for a DCM biomarker, offering valuable insights for early diagnosis and potential therapeutic development.
Collapse
Affiliation(s)
- Lingling Zhu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People’s Republic of China
| | - Ya Dong
- Department of Endocrinology, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Center for Diabetes Control and Prevention, Shenzhen, Guangdong Province, People’s Republic of China
| | - Hang Guo
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People’s Republic of China
| | - Jie Qiu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People’s Republic of China
| | - Jun Guo
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People’s Republic of China
| | - Yonghui Hu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Congqing Pan
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People’s Republic of China
| |
Collapse
|
8
|
Bianchi M, Kozyrev SV, Notarnicola A, Sandling JK, Pettersson M, Leonard D, Sjöwall C, Gunnarsson I, Rantapää‐Dahlqvist S, Bengtsson AA, Jönsen A, Svenungsson E, Enocsson H, Kvarnström M, Forsblad‐d'Elia H, Bucher SM, Norheim KB, Baecklund E, Jonsson R, Hammenfors D, Eriksson P, Mandl T, Omdal R, Padyukov L, Andersson H, Molberg Ø, Diederichsen LP, Syvänen A, Wahren‐Herlenius M, Nordmark G, Lundberg IE, Rönnblom L, Lindblad‐Toh K. Unraveling the Genetics of Shared Clinical and Serological Manifestations in Patients With Systemic Inflammatory Autoimmune Diseases. Arthritis Rheumatol 2025; 77:212-225. [PMID: 39284741 PMCID: PMC11782108 DOI: 10.1002/art.42988] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVE Systemic inflammatory autoimmune diseases (SIADs) such as systemic lupus erythematosus (SLE), primary Sjögren disease (pSS), and idiopathic inflammatory myopathies (myositis) are complex conditions characterized by shared circulating autoantibodies and clinical manifestations, including skin rashes, among others. This study was aimed at elucidating the genetics underlying these common features. METHODS We performed targeted DNA sequencing of coding and regulatory regions from approximately 1,900 immune-related genes in a large cohort of 2,292 well-characterized Scandinavian patients with SIADs with SLE, pSS, and myositis as well as 1,252 controls. A gene-based functionally weighted genetic score for aggregate testing of all genetic variants, including rare variants, was complemented by in silico functional analyses and in vitro reporter experiments. RESULTS Case-control association analysis detected known and potentially novel genetic loci in agreement with previous genetic and transcriptomics findings linked to the SIAD autoimmune background. Intriguingly, case-case comparisons between patient subgroups with and without specific autoantibodies revealed that the subgroups defined by antinuclear antibodies and anti-double-stranded DNA antibodies have unique genetic profiles reflecting their heterogeneity. When focusing on clinical features, we overall showed that dual-specificity phosphatase 1 (DUSP1) protective genetic variants lead to increased gene expression and potentially to anti-inflammatory effects on the SIAD-associated skin phenotype. This is consistent with recent genetic findings on eczema and with the previously reported down-regulation of the MAPK signaling-related gene DUSP1 in other skin disorders. CONCLUSION Together, this suggests common molecular mechanisms potentially underlying overlapping clinical manifestations shared among different disorders and informs clinical heterogeneity, which could be translated to improve disease diagnostic and treatment, also in more generalized disease frameworks.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Iva Gunnarsson
- Karolinska Institutet and Karolinska University HospitalStockholmSweden
| | | | | | | | | | | | - Marika Kvarnström
- Karolinska Institutet and Karolinska University HospitalStockholmSweden
| | | | | | | | | | | | | | | | | | - Roald Omdal
- Stavanger University Hospital, Stavanger, Norway, and University of BergenBergenNorway
| | - Leonid Padyukov
- Karolinska Institutet and Karolinska University HospitalStockholmSweden
| | | | | | - Louise Pyndt Diederichsen
- Odense University Hospital, Odense, Denmark, and Copenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | | | - Marie Wahren‐Herlenius
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden, and Broegelmann Research Laboratory, University of BergenBergenNorway
| | | | | | | | - Kerstin Lindblad‐Toh
- Uppsala University, Uppsala, Sweden, and Broad Institute of Massachusetts Institute of Technology and HarvardCambridgeMassachusetts
| |
Collapse
|
9
|
Zhao H, Mu Y, Liang A, Wei J, Lai S, Li X, Chen P, Li H, He H, Liu X, Liu H. Suppressing DUSP16 overexpression induced by ELK1 promotes neural progenitor cell differentiation in mouse models of Alzheimer's disease. Aging Cell 2025; 24:e14372. [PMID: 39434411 PMCID: PMC11822628 DOI: 10.1111/acel.14372] [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: 04/05/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Emerged evidence indicated that stimulating hippocampal neurogenesis is a potential strategy for restoring cognition in AD. Mitogen-activated protein kinases (MAPKs) play an essential role in neurogenesis. Meanwhile, the enzymatic power of the phosphatases is much greater than that of kinases. Dual-specificity phosphatase 16 (DUSP16), known to as a phosphatase negatively regulate MAPKs, may be implicated in neural differentiation. Nevertheless, the effect of DUSP16 on cognitive disorders by stimulating neural progenitor cell (NPC) differentiation in AD mice remains unclear. Our study demonstrates an association between DUSP16 SNPs and clinical progression in individuals with mild cognitive impairment (MCI). Besides, increased DUSP16 expression was detected in both 3xTg and SAMP8 mouse models of AD, accompanied by NPC neural differentiation impairments. By silencing DUSP16, the induction of neural differentiation, synaptic transmission, and cognitive benefits were observed in both AD mice. Furthermore, DUSP16 was involved in the process of NPC differentiation through regulating c-Jun N-terminal kinase (JNK) phosphorylation and SOX2 expression. Moreover, ETS transcription factor (ELK1) was involved in the DUSP16 transcription, which resulted in the upregulation of DUSP16 at the state of AD. Our data uncovers a potential regulatory role for DUSP16 in adult hippocampal neurogenesis (AHN) and provides a possibility to find a novel strategy for AD intervention.
Collapse
Affiliation(s)
- Huimin Zhao
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Yao Mu
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Anqi Liang
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Jie Wei
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Sixian Lai
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Xin Li
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Peipei Chen
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Hao Li
- Acupuncture and Moxibustion DepartmentJiangsu Provincial Second Chinese Medicine Hospital/the Second Affiliated Hospital of Nanjing University of Chinese MedicineNanjingChina
| | - Hua He
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Xiaoquan Liu
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| | - Haochen Liu
- Center of Drug Metabolism and PharmacokineticsChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
10
|
Morice A, de La Seiglière A, Kany A, Khonsari RH, Bensidhoum M, Puig-Lombardi ME, Legeai Mallet L. FGFR antagonists restore defective mandibular bone repair in a mouse model of osteochondrodysplasia. Bone Res 2025; 13:12. [PMID: 39837840 PMCID: PMC11751307 DOI: 10.1038/s41413-024-00385-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 01/30/2025] Open
Abstract
Gain-of-function mutations in fibroblast growth factor receptor (FGFR) genes lead to chondrodysplasia and craniosynostoses. FGFR signaling has a key role in the formation and repair of the craniofacial skeleton. Here, we analyzed the impact of Fgfr2- and Fgfr3-activating mutations on mandibular bone formation and endochondral bone repair after non-stabilized mandibular fractures in mouse models of Crouzon syndrome (Crz) and hypochondroplasia (Hch). Bone mineralization of the calluses was abnormally high in Crz mice and abnormally low in Hch mice. The latter model presented pseudarthrosis and impaired chondrocyte differentiation. Spatial transcriptomic analyses of the Hch callus revealed abnormally low expression of Col11, Col1a, Dmp1 genes in mature chondrocytes. We found that the expression of genes involved in autophagy and apoptosis (Smad1, Comp, Birc2) was significantly perturbed and that the Dusp3, Dusp9, and Socs3 genes controlling the mitogen-activated protein kinase pathway were overexpressed. Lastly, we found that treatment with a tyrosine kinase inhibitor (BGJ398, infigratinib) or a C-type natriuretic peptide (BMN111, vosoritide) fully rescued the defective endochondral bone repair observed in Hch mice. Taken as a whole, our findings show that FGFR3 is a critical orchestrator of bone repair and provide a rationale for the development of potential treatments for patients with FGFR3-osteochondrodysplasia.
Collapse
Affiliation(s)
- Anne Morice
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Paris, France
| | - Amélie de La Seiglière
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Paris, France
| | - Alexia Kany
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Paris, France
| | - Roman H Khonsari
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Paris, France
| | | | - Maria-Emilia Puig-Lombardi
- Bioinformatics Core Platform, Imagine Institute, INSERM UMR1163 and Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR3633, Université Paris Cité, Paris, France
| | - Laurence Legeai Mallet
- Université de Paris Cité, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Paris, France.
| |
Collapse
|
11
|
He Z, Uto T, Tanigawa S, Sakao K, Kumamoto T, Xie K, Pan X, Wu S, Yang Y, Komatsu M, Hou D. Fisetin is a selective adenosine triphosphate-competitive inhibitor for mitogen-activated protein kinase kinase 4 to inhibit lipopolysaccharide-stimulated inflammation. Biofactors 2025; 51:e2108. [PMID: 39087587 PMCID: PMC11680972 DOI: 10.1002/biof.2108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
The mitogen-activated protein kinase kinase 4 (MKK4), a member of the MAP kinase kinase family, directly phosphorylates and activates the c-Jun NH2-terminal kinases (JNK), in response to proinflammatory cytokines and cellular stresses. Regulation of the MKK4 activity is considered to be a novel approach for the prevention and treatment of inflammation. The aim of this study was to identify whether fisetin, a potential anti-inflammatory compound, targets MKK4-JNK cascade to inhibit lipopolysaccharide (LPS)-stimulated inflammatory response. RAW264 macrophage pretreated with fisetin following LPS stimulation was used as a cell model to investigate the transactivation and expression of related-inflammatory genes by transient transfection assay, electrophoretic mobility shift assay (EMSA), or enzyme-linked immunosorbent assay (ELISA), and cellular signaling as well as binding of related-signal proteins by Western blot, pull-down assay and kinase assay, and molecular modeling. The transactivation and expression of cyclooxygenase-2 (COX-2) gene as well as prostaglandin E2 (PGE2) secretion induced by LPS were inhibited by fisetin in a dose-dependent manner. Signaling transduction analysis demonstrated that fisetin selectively inhibited MKK4-JNK1/2 signaling to suppress the phosphorylation of transcription factor AP-1 without affecting the NF-κB and Jak2-Stat3 signaling as well as the phosphorylation of Src, Syk, and TAK1. Furthermore, in vitro and ex vivo pull-down assay using cell lysate or purified protein demonstrated that fisetin could bind directly to MKK4. Molecular modeling using the Molecular Operating Environment™ software indicated that fisetin docked into the ATP-binding pocket of MKK4 with a binding energy of -71.75 kcal/mol and formed a 1.70 Å hydrogen bound with Asp247 residue of MKK4. The IC50 of fisetin against MKK4 was estimated as 2.899 μM in the kinase assay, and the ATP-competitive effect was confirmed by ATP titration. Taken together, our data revealed that fisetin is a potent selective ATP-competitive MKK4 inhibitor to suppress MKK4-JNK1/2-AP-1 cascade for inhibiting LPS-induced inflammation.
Collapse
Affiliation(s)
- Ziyu He
- The United Graduate School of Agricultural SciencesKagoshima UniversityKagoshimaJapan
| | - Takuhiro Uto
- Department of Pharmacy, Faculty of Pharmaceutical SciencesNagasaki International UniversitySaseboJapan
| | - Shunsuke Tanigawa
- Department of Kidney Development, Institute of Molecular Embryology and GeneticsKumamoto UniversityKumamotoJapan
| | - Kozue Sakao
- The United Graduate School of Agricultural SciencesKagoshima UniversityKagoshimaJapan
- Graduate School of Agriculture, Forestry and FisheriesKagoshima UniversityKagoshimaJapan
| | - Takuma Kumamoto
- Department of Brain & NeurosciencesTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Kun Xie
- The United Graduate School of Agricultural SciencesKagoshima UniversityKagoshimaJapan
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and TechnologyHunan Agricultural UniversityChangshaPeople's Republic of China
| | - Xuchi Pan
- Graduate School of Agriculture, Forestry and FisheriesKagoshima UniversityKagoshimaJapan
| | - Shusong Wu
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and TechnologyHunan Agricultural UniversityChangshaPeople's Republic of China
| | - Yili Yang
- China Regional Research CentreInternational Centre for Genetic Engineering and BiotechnologyTaizhouPeople's Republic of China
| | - Masaharu Komatsu
- The United Graduate School of Agricultural SciencesKagoshima UniversityKagoshimaJapan
- Graduate School of Agriculture, Forestry and FisheriesKagoshima UniversityKagoshimaJapan
| | - De‐Xing Hou
- The United Graduate School of Agricultural SciencesKagoshima UniversityKagoshimaJapan
- Graduate School of Agriculture, Forestry and FisheriesKagoshima UniversityKagoshimaJapan
| |
Collapse
|
12
|
Wei B, Shi Y, Yu X, Cai Y, Zhao Y, Song Y, Zhao Z, Huo M, Li L, Gao Q, Yu D, Wang B, Sun M. GR/P300 Regulates MKP1 Signaling Pathway and Mediates Depression-like Behavior in Prenatally Stressed Offspring. Mol Neurobiol 2024; 61:10613-10628. [PMID: 38769227 DOI: 10.1007/s12035-024-04244-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Accumulating evidence suggests that prenatal stress (PNS) increases offspring susceptibility to depression, but the underlying mechanisms remain unclear. We constructed a mouse model of prenatal stress by spatially restraining pregnant mice from 09:00-11:00 daily on Days 5-20 of gestation. In this study, western blot analysis, quantitative real-time PCR (qRT‒PCR), immunofluorescence, immunoprecipitation, chromatin immunoprecipitation (ChIP), and mifepristone rescue assays were used to investigate alterations in the GR/P300-MKP1 and downstream ERK/CREB/TRKB pathways in the brains of prenatally stressed offspring to determine the pathogenesis of the reduced neurogenesis and depression-like behaviors in offspring induced by PNS. We found that prenatal stress leads to reduced hippocampal neurogenesis and depression-like behavior in offspring. Prenatal stress causes high levels of glucocorticoids to enter the fetus and activate the hypothalamic‒pituitary‒adrenal (HPA) axis, resulting in decreased hippocampal glucocorticoid receptor (GR) levels in offspring. Furthermore, the nuclear translocation of GR and P300 (an acetylation modifying enzyme) complex in the hippocampus of PNS offspring increased significantly. This GR/P300 complex upregulates MKP1, which is a negative regulator of the ERK/CREB/TRKB signaling pathway associated with depression. Interestingly, treatment with a GR antagonist (mifepristone, RU486) increased hippocampal GR levels and decreased MKP1 expression, thereby ameliorating abnormal neurogenesis and depression-like behavior in PNS offspring. In conclusion, our study suggested that the regulation of the MKP1 signaling pathway by GR/P300 is involved in depression-like behavior in prenatal stress-exposed offspring and provides new insights and ideas for the fetal hypothesis of mental health.
Collapse
Affiliation(s)
- Bin Wei
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250000, Shandong, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yajun Shi
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Xi Yu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yongle Cai
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yan Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yueyang Song
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Zejun Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Ming Huo
- Reproductive Medicine Center, The First Hospital of Lanzhou University, LanzhouGansu, 730000, China
| | - Lingjun Li
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Qinqin Gao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Dongyi Yu
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250000, Shandong, China
| | - Bin Wang
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China.
| | - Miao Sun
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, 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.
| |
Collapse
|
13
|
Ma H, Gao G, Palti Y, Tripathi V, Birkett JE, Weber GM. Transcriptomic Response of the Ovarian Follicle Complex in Post-Vitellogenic Rainbow Trout to 17α,20β-Dihdroxy-4-pregnen-3-one In Vitro. Int J Mol Sci 2024; 25:12683. [PMID: 39684392 DOI: 10.3390/ijms252312683] [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: 11/07/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Gonadotropins and progestins are the primary regulators of follicle maturation and ovulation in fish, and they require complex communication among the oocyte and somatic cells of the follicle. The major progestin and the maturation-inducing hormone in salmonids is 17α,20β-dihdroxy-4-pregnen-3-one (17,20βP), and traditional nuclear receptors and membrane steroid receptors for the progestin have been identified within the follicle. Herein, RNA-seq was used to conduct a comprehensive survey of changes in gene expression throughout the intact follicle in response to in vitro treatment with these hormones to provide a foundation for understanding the coordination of their actions in regulating follicle maturation and preparation for ovulation. A total of 5292 differentially expressed genes were identified from our transcriptome sequencing datasets comparing four treatments: fresh tissue; untreated control; 17,20βP-treated; and salmon pituitary homogenate-treated follicles. Extensive overlap in affected genes suggests many gonadotropin actions leading to the acquisition of maturational and ovulatory competence are mediated in part by gonadotropin induction of 17,20βP synthesis. KEGG analysis identified signaling pathways, including MAPK, TGFβ, FoxO, and Wnt signaling pathways, among the most significantly enriched pathways altered by 17,20βP treatment, suggesting pervasive influences of 17,20βP on actions of other endocrine and paracrine factors in the follicle complex.
Collapse
Affiliation(s)
- Hao Ma
- US Department of Agriculture, Agricultural Research Service, National Animal Disease Center, Ruminant Disease and Immunology Research Unit, Ames, IA 50010, USA
| | - Guangtu Gao
- US Department of Agriculture, Agricultural Research Service, National Center for Cool and Cold Water Aquaculture, 11861 Leetown Road, Kearneysville, WV 25430, USA
| | - Yniv Palti
- US Department of Agriculture, Agricultural Research Service, National Center for Cool and Cold Water Aquaculture, 11861 Leetown Road, Kearneysville, WV 25430, USA
| | - Vibha Tripathi
- US Department of Agriculture, Agricultural Research Service, National Center for Cool and Cold Water Aquaculture, 11861 Leetown Road, Kearneysville, WV 25430, USA
| | - Jill E Birkett
- US Department of Agriculture, Agricultural Research Service, National Center for Cool and Cold Water Aquaculture, 11861 Leetown Road, Kearneysville, WV 25430, USA
| | - Gregory M Weber
- US Department of Agriculture, Agricultural Research Service, National Center for Cool and Cold Water Aquaculture, 11861 Leetown Road, Kearneysville, WV 25430, USA
| |
Collapse
|
14
|
Zheng L, Wang J, Han S, Zhong L, Liu Z, Li B, Zhang R, Zhou L, Zheng X, Liu Z, Zeng C, Li R, Zou Y, Wang L, Wu Y, Kang T. The KLF16/MYC feedback loop is a therapeutic target in bladder cancer. J Exp Clin Cancer Res 2024; 43:303. [PMID: 39551759 PMCID: PMC11571712 DOI: 10.1186/s13046-024-03224-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Bladder cancer (BLCA) is a common malignancy characterized by dysregulated transcription and a lack of effective therapeutic targets. In this study, we aimed to identify and evaluate novel targets with clinical potential essential for tumor growth in BLCA. METHODS CRISPR-Cas9 screening was used to identify transcription factors essential for bladder cancer cell viability. The biological functions of KLF16 in bladder cancer were investigated both in vitro and in vivo. The regulatory mechanism between KLF16 and MYC was elucidated through a series of analyses, including RNA sequencing, quantitative polymerase chain reaction (qPCR), RNA immunoprecipitation, Western blotting, Mass spectrometry, Dual-luciferase reporter assays, Cleavage Under Targets and Tagmentation (CUT&Tag) sequencing, OptoDroplets assays, and RNA stability assay. The clinical relevance of KLF16 and MYC in bladder cancer was evaluated through analyses of public databases and immunohistochemistry. RESULTS Krüppel-like factor 16 (KLF16) was essential for BLCA cell viability. Elevated expression of KLF16 was observed in bladder cancer tissues, and higher expression levels of KLF16 were correlated with poor progression-free survival (PFS) and cancer-specific survival (CSS) probabilities in BLCA patients. Mechanistically, KLF16 mRNA competed with the mRNA of dual-specificity phosphatase 16 (DUSP16) for binding to the RNA-binding protein, WW domain binding protein 11 (WBP11), resulting in destabilization of the DUSP16 mRNA. This, in turn, led to activation of ERK1/2, which stabilized the MYC protein. Furthermore, KLF16 interacted with MYC to form nuclear condensates, thereby enhancing MYC's transcriptional activity. Additionally, MYC transcriptionally upregulated KLF16, creating a positive feedback loop between KLF16 and MYC that amplified their oncogenic functions. Targeting this loop with bromodomain inhibitors, such as OTX015 and ABBV-744, suppressed the transcription of both KLF16 and MYC, resulting in reduced BLCA cell viability and tumor growth, as well as increased sensitivity to chemotherapy. CONCLUSIONS Our study revealed the crucial role of the KLF16/MYC regulatory axis in modulating tumor growth and chemotherapy sensitivity in BLCA, suggesting that combining bromodomain inhibitors, such as OTX015 or ABBV-744, with DDP or gemcitabine could be a promising therapeutic intervention for BLCA patients.
Collapse
Affiliation(s)
- Lisi Zheng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Jingxuan Wang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Shan Han
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Li Zhong
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
- Center of Digestive Disease, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, People's Republic of China
| | - Zefu Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Bin Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Ruhua Zhang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Liwen Zhou
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Xianchong Zheng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Zhenhua Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Cuiling Zeng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Ruonan Li
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Yezi Zou
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Liqin Wang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| | - Yuanzhong Wu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| | - Tiebang Kang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
15
|
Guo S, Zeng M, Zhang C, Fan Y, Ran M, Song Z. Genome-wide characterization and comparative expression profiling of dual-specificity phosphatase genes in yellow catfish ( Pelteobagrus fulvidraco) after infection with exogenous Aeromonas hydrophila. Front Immunol 2024; 15:1481696. [PMID: 39606227 PMCID: PMC11598348 DOI: 10.3389/fimmu.2024.1481696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/15/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Dual-specificity phosphatases (DUSPs) are crucial regulators in many mammals, managing dephosphorylation and inactivation of mitogen-activated protein kinases (MAPKs) and playing essential roles in immune responses. However, their presence and functions in teleosts, like the yellow catfish (Pelteobagrus fulvidraco), remain unexplored. Methods In this study, eight pfDusp genes (pfDusp1-7 and pfDusp10) were identified in yellow catfish. We characterized their molecular features, conserved protein sequences, and chromosomal localization through genome-wide analyses, and we examined their expression patterns in immune responses. Results Our findings reveal two conserved motifs, Leu-Phe-Leu-Gly and Ala-Tyr-Leu-Met, within the DSPc domain of DUSP proteins. The genes were mapped across seven chromosomes without evidence of duplication. Comparative analysis showed high conservation of Dusp genes across vertebrates, with evolutionary analysis suggesting Dusp3 as a potential intermediate form. Dusp transcripts were significantly upregulated in the kidney post-A. hydrophila infection. Discussion These results suggest the involvement of Dusp genes in the immune response of yellow catfish to bacterial pathogens, providing insights into their evolutionary significance and potential applications in aquaculture and molecular breeding.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhaobin Song
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College
of Life Sciences, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Arora R, Malla WA, Tyagi A, Saxena S, Mahajan S, Sajjanar B, Gandham R, Tiwari AK. Transcriptome profiling of Canine Parvovirus 2 Nonstructural gene 1(CPV2.NS1) transfected 4T1 mice mammary tumor cells to elucidate its oncolytic effects. Int J Biol Macromol 2024; 281:136620. [PMID: 39419151 DOI: 10.1016/j.ijbiomac.2024.136620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
Oncolytic viral gene therapy is a directed approach to target cancer cells without affecting healthy cells of the body. Canine parvovirus (CPV2) is an oncolytic virus that precisely targets and destroys neoplastic cells by causing DNA damage, mitochondrial damage, and apoptosis. Non-structural gene 1 (NS1) of CPV, concerned with viral DNA replication is a key mediator of cytotoxicity of CPV and can specifically cause tumor cell lysis. In the present study, by using the transcriptomics approach, we tried to identify molecular pathways and key genes involved in CPV2.NS1 mediated 4T1 mice mammary tumor cell death. We identified necroptosis and mitochondrial damage-mediated apoptosis as major cell death pathways leading to CPV2.NS1 transfected 4T1 cancer cell death. Various DEGs identified in our study play an important role in pathways like the PI3K/AKT pathway, diverse metabolic pathways, MAPK signaling pathway, and FGF signaling pathway, whichare mostly dysregulated in cancerous conditions. Histone variant H2A.X genes, Capn2, and Mapk10/JNK are predicted as key genes that play a role in causing endoplasmic reticulum stress and mitochondrial damage, thereby leading to necroptosis and apoptosis. This study is a preliminary work done to identify key genes and molecular pathways involved in CPV2.NS1 mediated 4T1 cancer cells death which need to be further validated to establish this viral gene as a potent oncolytic agent.
Collapse
Affiliation(s)
- Richa Arora
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India; Department of Veterinary Biochemistry, College of Veterinary and Animal Sciences, Kishanganj, BASU, Patna, India
| | - Waseem Akram Malla
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Arpit Tyagi
- GB Pant University of Agriculture and Technology, Pantnagar, India
| | - Shikha Saxena
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Sonalika Mahajan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Basavaraj Sajjanar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Ravikumar Gandham
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | | |
Collapse
|
17
|
Pan Z, Yao Y, Liu X, Wang Y, Zhang X, Zha S, Hu K. Nr1d1 inhibition mitigates intermittent hypoxia-induced pulmonary hypertension via Dusp1-mediated Erk1/2 deactivation and mitochondrial fission attenuation. Cell Death Discov 2024; 10:459. [PMID: 39472573 PMCID: PMC11522549 DOI: 10.1038/s41420-024-02219-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 09/30/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
Intermittent hypoxia (IH) precipitates pulmonary vasoconstriction, culminating in the onset of pulmonary hypertension (PH) among individuals afflicted with sleep apnea. While Nuclear receptor subfamily 1 group D member 1 (Nr1d1) is progressively recognized as pivotal regulator of cellular physiology, the role in the pathogenesis of IH-induced PH remains largely uncharted. The expression of Nr1d1 was examined in IH-induced rodent PH and in IH-treated PASMCs. To elucidate the contribution of Nr1d1 to the development of IH-induced PH, we employed siRNA to modulate Nr1d1 expression in vitro and employed serotype 1 adeno-associated virus (AAV1) in vivo. Nr1d1 levels were elevated in IH-induced rodents PH lung tissues and IH-treated PASMCs. Knocking down Nr1d1 by AAV1 effectively inhibited PH progression in chronic IH-induced PH models. Mechanistic investigations identified dual specificity phosphatase 1 (Dusp1), as a direct target that Nr1d1 trans-repressed, mediating Nr1d1's regulatory influence on Erk1/2/Drp1 signaling. Nr1d1 deficiency ameliorates mitochondrial dysfunction and fission by restoring Dusp1 dysregulation and Drp1 phosphorylation. Activation of Erk1/2 with PMA reversed the Dusp1-mediated regulation of Drp1 phosphorylation, indicating the involvement of the Erk1/2 pathway in Drp1 phosphorylation controlled by Dusp1. Meanwhile, intermittent hypoxia induced more severe PH in Dusp1 knockout mice compared with wild-type mice. Our data unveil a novel role for Nr1d1 in IH-induced PH pathogenesis and an undisclosed Nr1d1-Dusp1 axis in PASMCs mitochondrial fission regulation.
Collapse
Affiliation(s)
- Zhou Pan
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Yao
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xu Liu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yixuan Wang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinyue Zhang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shiqian Zha
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Hu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
18
|
Li N, Yun B, Zeng L, Lv Y, Zhou Y, Fang M, Li S, Chen Y, Huang E, Zhang L, Jiang Y, Zhang H, Li J, Yuan X. The antisense lncRNA of TAB2 that prevents oxidative stress to enhance the follicular growth in mammals. Commun Biol 2024; 7:1246. [PMID: 39358475 PMCID: PMC11447032 DOI: 10.1038/s42003-024-06960-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
LncRNAs are highly implicated in oxidative stress (OS) during the growth of mammalian follicles. TAK1 binding protein 2 gene (TAB2) has been suggested to involve in the normal apoptosis and proliferation of granulosa cells (GCs), the main supporting cells in ovarian follicles. In this study, we found that TAB2 increased the expressions of SOD1, P50, and P65 to suppress the OS, thereby inhibiting the apoptosis and promoting the proliferation in GCs. Notably, DNMTs appeared to mediate the expression of TAB2 without the changes of DNA methylation at TAB2's promoter. We identified an antisense lncRNA of TAB2, discovered that DNA methylation regulated the transcription of TAB2-AS in GCs, and found TAB2-AS medicated the follicular growth of ovaries in vivo. Mechanistically, the hypomethylation of the CpG site (-1759/-1760) activated the transcription of TAB2-AS, and the 1-155 nt and 156-241 nt of TAB2-AS were respectively complementary to 4368-4534 nt and 4215-4300 nt of TAB2's mRNA to increase the expression of TAB2. Moreover, TAB2-AS inhibited the OS and apoptosis of GCs, while promoted the proliferation of GCs to expedite the follicular growth, which was in line with that of TAB2. Collectively, these findings revealed the antisense lncRNA mechanism mediated by DNA methylation, and TAB2-AS might be the target to control OS during follicular growth in mammals.
Collapse
Affiliation(s)
- Nian Li
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Bing Yun
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Liqing Zeng
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Yuanyuan Lv
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Yinqi Zhou
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Ming Fang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Shuo Li
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Yongcai Chen
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Enyuan Huang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Liuhong Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Yao Jiang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, 6149, Australia
| | - Hao Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jiaqi Li
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| | - Xiaolong Yuan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, 6150, Australia.
| |
Collapse
|
19
|
Zeng Y, Bai X, Zhu G, Zhu M, Peng W, Song J, Cai H, Ye L, Chen C, Song Y, Jin M, Zhang XQ, Wang J. m 6A-mediated HDAC9 upregulation promotes particulate matter-induced airway inflammation via epigenetic control of DUSP9-MAPK axis and acts as an inhaled nanotherapeutic target. JOURNAL OF HAZARDOUS MATERIALS 2024; 477:135093. [PMID: 39088948 DOI: 10.1016/j.jhazmat.2024.135093] [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: 03/12/2024] [Revised: 06/15/2024] [Accepted: 07/01/2024] [Indexed: 08/03/2024]
Abstract
Exposure to particulate matter (PM) can cause airway inflammation and worsen various airway diseases. However, the underlying molecular mechanism by which PM triggers airway inflammation has not been completely elucidated, and effective interventions are lacking. Our study revealed that PM exposure increased the expression of histone deacetylase 9 (HDAC9) in human bronchial epithelial cells and mouse airway epithelium through the METTL3/m6A methylation/IGF2BP3 pathway. Functional assays showed that HDAC9 upregulation promoted PM-induced airway inflammation and activation of MAPK signaling pathway in vitro and in vivo. Mechanistically, HDAC9 modulated the deacetylation of histone 4 acetylation at K12 (H4K12) in the promoter region of dual specificity phosphatase 9 (DUSP9) to repress the expression of DUSP9 and resulting in the activation of MAPK signaling pathway, thereby promoting PM-induced airway inflammation. Additionally, HDAC9 bound to MEF2A to weaken its anti-inflammatory effect on PM-induced airway inflammation. Then, we developed a novel inhaled lipid nanoparticle system for delivering HDAC9 siRNA to the airway, offering an effective treatment for PM-induced airway inflammation. Collectively, we elucidated the crucial regulatory mechanism of HDAC9 in PM-induced airway inflammation and introduced an inhaled therapeutic approach targeting HDAC9. These findings contribute to alleviating the burden of various airway diseases caused by PM exposure.
Collapse
Affiliation(s)
- Yingying Zeng
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Bai
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guiping Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mengchan Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenjun Peng
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Juan Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hui Cai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ling Ye
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cuicui Chen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai 200032, China; Shanghai Respiratory Research Institute, Shanghai 200032, China
| | - Meiling Jin
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xue-Qing Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jian Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
20
|
Skeens E, Maschietto F, Manjula R, Shillingford S, Lolis EJ, Batista VS, Bennett AM, Lisi GP. Dynamic and structural insights into allosteric regulation on MKP5 a dual-specificity phosphatase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611520. [PMID: 39282375 PMCID: PMC11398491 DOI: 10.1101/2024.09.05.611520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Dual-specificity mitogen-activated protein kinase (MAPK) phosphatases (MKPs) directly dephosphorylate and inactivate the MAPKs. Although the catalytic mechanism of dephosphorylation of the MAPKs by the MKPs is established, a complete molecular picture of the regulatory interplay between the MAPKs and MKPs still remains to be fully explored. Here, we sought to define the molecular mechanism of MKP5 regulation through an allosteric site within its catalytic domain. We demonstrate using crystallographic and NMR spectroscopy approaches that residue Y435 is required to maintain the structural integrity of the allosteric pocket. Along with molecular dynamics simulations, these data provide insight into how changes in the allosteric pocket propagate conformational flexibility in the surrounding loops to reorganize catalytically crucial residues in the active site. Furthermore, Y435 contributes to the interaction with p38 MAPK and JNK, thereby promoting dephosphorylation. Collectively, these results highlight the role of Y435 in the allosteric site as a novel mode of MKP5 regulation by p38 MAPK and JNK.
Collapse
Affiliation(s)
- Erin Skeens
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | | | - Ramu Manjula
- Department of Pharmacology, Yale School of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Shanelle Shillingford
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
- Department of Pharmacology, Yale School of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Elias J. Lolis
- Department of Pharmacology, Yale School of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Victor S. Batista
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
| | - Anton M. Bennett
- Department of Pharmacology, Yale School of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| | - George P. Lisi
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
21
|
Fish JL. Fibroblast growth factors-An introduction to our primer series. Differentiation 2024; 139:100804. [PMID: 39117549 DOI: 10.1016/j.diff.2024.100804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Affiliation(s)
- Jennifer L Fish
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, USA.
| |
Collapse
|
22
|
Sun Y, Zhou R, Hu J, Feng S, Hu Q. Reversible control of kinase signaling through chemical-induced dephosphorylation. Commun Biol 2024; 7:1073. [PMID: 39217250 PMCID: PMC11366001 DOI: 10.1038/s42003-024-06771-9] [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: 12/24/2023] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The coordination between kinases and phosphatases is crucial for regulating the phosphorylation levels of essential signaling molecules. Methods enabling precise control of kinase activities are valuable for understanding the kinase functions and for developing targeted therapies. Here, we use the abscisic acid (ABA)-induced proximity system to reversibly control kinase signaling by recruiting phosphatases. Using this method, we found that the oncogenic tyrosine kinase BCR::ABL1 can be inhibited by recruiting various cytoplasmic phosphatases. We also discovered that the oncogenic serine/threonine kinase BRAF(V600E), which has been reported to bypass phosphorylation regulation, can be positively regulated by protein phosphatase 1 (PP1) and negatively regulated by PP5. Additionally, we observed that the dual-specificity kinase MEK1 can be inhibited by recruiting PP5. This suggests that bifunctional molecules capable of recruiting PP5 to MEK or RAF kinases could be promising anticancer drug candidates. Thus, the ABA-induced dephosphorylation method enables rapid screening of phosphatases to precisely control kinase signaling.
Collapse
Affiliation(s)
- Ying Sun
- Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Rihong Zhou
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jin Hu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Shan Feng
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Qi Hu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
23
|
Su J, Song Y, Zhu Z, Huang X, Fan J, Qiao J, Mao F. Cell-cell communication: new insights and clinical implications. Signal Transduct Target Ther 2024; 9:196. [PMID: 39107318 PMCID: PMC11382761 DOI: 10.1038/s41392-024-01888-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/09/2024] [Accepted: 06/02/2024] [Indexed: 09/11/2024] Open
Abstract
Multicellular organisms are composed of diverse cell types that must coordinate their behaviors through communication. Cell-cell communication (CCC) is essential for growth, development, differentiation, tissue and organ formation, maintenance, and physiological regulation. Cells communicate through direct contact or at a distance using ligand-receptor interactions. So cellular communication encompasses two essential processes: cell signal conduction for generation and intercellular transmission of signals, and cell signal transduction for reception and procession of signals. Deciphering intercellular communication networks is critical for understanding cell differentiation, development, and metabolism. First, we comprehensively review the historical milestones in CCC studies, followed by a detailed description of the mechanisms of signal molecule transmission and the importance of the main signaling pathways they mediate in maintaining biological functions. Then we systematically introduce a series of human diseases caused by abnormalities in cell communication and their progress in clinical applications. Finally, we summarize various methods for monitoring cell interactions, including cell imaging, proximity-based chemical labeling, mechanical force analysis, downstream analysis strategies, and single-cell technologies. These methods aim to illustrate how biological functions depend on these interactions and the complexity of their regulatory signaling pathways to regulate crucial physiological processes, including tissue homeostasis, cell development, and immune responses in diseases. In addition, this review enhances our understanding of the biological processes that occur after cell-cell binding, highlighting its application in discovering new therapeutic targets and biomarkers related to precision medicine. This collective understanding provides a foundation for developing new targeted drugs and personalized treatments.
Collapse
Affiliation(s)
- Jimeng Su
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ying Song
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Zhipeng Zhu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Xinyue Huang
- Biomedical Research Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
24
|
Chen Z, Wang C, Huang S, Shi Y, Xi R. Directly selecting cell-type marker genes for single-cell clustering analyses. CELL REPORTS METHODS 2024; 4:100810. [PMID: 38981475 PMCID: PMC11294843 DOI: 10.1016/j.crmeth.2024.100810] [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: 11/14/2023] [Revised: 03/16/2024] [Accepted: 06/12/2024] [Indexed: 07/11/2024]
Abstract
In single-cell RNA sequencing (scRNA-seq) studies, cell types and their marker genes are often identified by clustering and differentially expressed gene (DEG) analysis. A common practice is to select genes using surrogate criteria such as variance and deviance, then cluster them using selected genes and detect markers by DEG analysis assuming known cell types. The surrogate criteria can miss important genes or select unimportant genes, while DEG analysis has the selection-bias problem. We present Festem, a statistical method for the direct selection of cell-type markers for downstream clustering. Festem distinguishes marker genes with heterogeneous distribution across cells that are cluster informative. Simulation and scRNA-seq applications demonstrate that Festem can sensitively select markers with high precision and enables the identification of cell types often missed by other methods. In a large intrahepatic cholangiocarcinoma dataset, we identify diverse CD8+ T cell types and potential prognostic marker genes.
Collapse
Affiliation(s)
- Zihao Chen
- School of Mathematical Sciences and Center for Statistical Science, Peking University, Beijing 100871, China
| | - Changhu Wang
- School of Mathematical Sciences and Center for Statistical Science, Peking University, Beijing 100871, China
| | - Siyuan Huang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yang Shi
- BeiGene (Beijing) Co., Ltd., Beijing 100871, China
| | - Ruibin Xi
- School of Mathematical Sciences and Center for Statistical Science, Peking University, Beijing 100871, China.
| |
Collapse
|
25
|
Zhang P, Zheng J, Han T, Ma J, Gnanashanmugam D, Li M, Tang YW, Deng G. A blood-based 3-gene signature score for therapeutic monitoring in patients with pulmonary tuberculosis. Tuberculosis (Edinb) 2024; 147:102521. [PMID: 38801793 DOI: 10.1016/j.tube.2024.102521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/12/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVE To assess the validity of Xpert Tuberculosis Fingerstick score for monitoring treatment response and analyze factors influencing its performance. METHODS 122 adults with pulmonary tuberculosis were recruited and stratified into three cohorts: Diabetic-drug-susceptible-TB (DM-TB), Non-diabetic-drug-susceptible-TB (NDM-TB) and Non-diabetic Multidrug-resistant TB (MDR-TB). Fingerstick blood specimens were tested at treatment initiation (M0) and the end of the first (M1), second (M2), and sixth month (M6) to generate a TB-score. RESULTS The TB-score in all participants yielded an AUC of 0.707 (95% CI: 0.579-0.834) at M2 when its performance was evaluated against sputum culture conversion. In all non-diabetes patients, the AUC reached 0.88 (95% CI: 0.756-1.000) with an optimal cut-off value of 1.95 at which sensitivity was 90.0% (95% CI: 59.6-98.2%) and specificity was 81.3% (95% CI: 70.0-88.9%). The mean TB score was higher in patients with low bacterial loads (n = 31) than those with high bacterial loads (n = 91) at M0, M1, M2, and M6, and was higher in non-cavitary patients (n = 71) than those with cavitary lesions (n = 51) at M0, M1, and M2. CONCLUSION Xpert TB-score shows promising predictive value for culture conversion in non-diabetic TB patients. Sputum bacterial load and lung cavitation status have an influence on the value of TB score.
Collapse
Affiliation(s)
- Peize Zhang
- Department of Pulmonary Medicine and Tuberculosis, The Third People's Hospital of Shenzhen, China.
| | - Junfeng Zheng
- Department of Pulmonary Medicine and Tuberculosis, The Third People's Hospital of Shenzhen, China.
| | - Tingting Han
- Guangdong Medical University, The First Clinical Medical College, Zhanjiang, Guangdong, China.
| | - Jian Ma
- Medical Affairs, Danaher Corporation/Cepheid (China), Shanghai, China.
| | | | - Mengran Li
- Department of Biostatistics & Data Management, Beckman Coulter, Shanghai, China.
| | - Yi-Wei Tang
- Medical Affairs, Danaher Corporation/Cepheid (China), Shanghai, China.
| | - Guofang Deng
- Department of Pulmonary Medicine and Tuberculosis, The Third People's Hospital of Shenzhen, China.
| |
Collapse
|
26
|
Reddi KK, Chava S, Chabattula SC, Edwards YJK, Singh K, Gupta R. ASAH1 facilitates TNBC by DUSP5 suppression-driven activation of MAP kinase pathway and represents a therapeutic vulnerability. Cell Death Dis 2024; 15:452. [PMID: 38926346 PMCID: PMC11208621 DOI: 10.1038/s41419-024-06831-2] [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: 12/28/2023] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is prone to metastasis and therapy resistance. Owing to its aggressive nature and limited availability of targeted therapies, TNBC is associated with higher mortality as compared to other forms of breast cancer. In order to develop new therapeutic options for TNBC, we characterized the factors involved in TNBC growth and progression. Here, we demonstrate that N-acylsphingosine amidohydrolase 1 (ASAH1) is overexpressed in TNBC cells and is regulated via p53 and PI3K-AKT signaling pathways. Genetic knockdown or pharmacological inhibition of ASAH1 suppresses TNBC growth and progression. Mechanistically, ASAH1 inhibition stimulates dual-specificity phosphatase 5 (DUSP5) expression, suppressing the mitogen-activated protein kinase (MAPK) pathway. Furthermore, pharmacological cotargeting of the ASAH1 and MAPK pathways inhibits TNBC growth. Collectively, we unmasked a novel role of ASAH1 in driving TNBC and identified dual targeting of the ASAH1 and MAPK pathways as a potential new therapeutic approach for TNBC treatment.
Collapse
Affiliation(s)
- Kiran Kumar Reddi
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suresh Chava
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Siva Chander Chabattula
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yvonne J K Edwards
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kamaljeet Singh
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center at The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
27
|
Liu K, Li Z, Li L, Heyward S, Wang SR, He L, Wang H. Mechanistic Understanding of Dexamethasone-Mediated Protection against Remdesivir-Induced Hepatotoxicity. Mol Pharmacol 2024; 106:71-82. [PMID: 38769019 DOI: 10.1124/molpharm.124.000894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/22/2024] Open
Abstract
Remdesivir (RDV), a broad-spectrum antiviral agent, is often used together with dexamethasone (DEX) for hospitalized COVID-19 patients requiring respiratory support. Potential hepatic adverse drug reaction is a safety concern associated with the use of RDV. We previously reported that DEX cotreatment effectively mitigates RDV-induced hepatotoxicity and reduces elevated serum alanine aminotransferase and aspartate aminotransferase levels in cultured human primary hepatocytes (HPH) and hospitalized COVID-19 patients, respectively. Yet, the precise mechanism behind this protective drug-drug interaction remains largely unknown. Here, we show that through the activation of p38, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinases 1 and 2 (ERK1/2) signaling, RDV induces apoptosis (cleavage of caspases 8, 9, and 3), autophagy (increased autophagosome and LC3-II), and mitochondrial damages (decreased membrane potential, respiration, ATP levels, and increased expression of Bax and the released cytosolic cytochrome C) in HPH. Importantly, cotreatment with DEX partially reversed RDV-induced apoptosis, autophagy, and cell death. Mechanistically, DEX deactivates/dephosphorylates p38, JNK, and ERK1/2 signaling by enhancing the expression of dual specificity protein phosphatase 1 (DUSP1), a mitogen-activated protein kinase (MAPK) phosphatase, in a glucocorticoid receptor (GR)-dependent manner. Knockdown of GR in HPH attenuates DEX-mediated DUSP1 induction, MAPK dephosphorylation, as well as protection against RDV-induced hepatotoxicity. Collectively, our findings suggest a molecular mechanism by which DEX modulates the GR-DUSP1-MAPK regulatory axis to alleviate the adverse actions of RDV in the liver. SIGNIFICANCE STATEMENT: The research uncovers the molecular mechanisms by which dexamethasone safeguards against remdesivir-associated liver damage in the context of COVID-19 treatment.
Collapse
Affiliation(s)
- Kaiyan Liu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Zhihui Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Scott Heyward
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Shelley R Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Ling He
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| |
Collapse
|
28
|
Kesarwani M, Kincaid Z, Azhar M, Azam M. Enhanced MAPK signaling induced by CSF3R mutants confers dependence to DUSP1 for leukemic transformation. Blood Adv 2024; 8:2765-2776. [PMID: 38531054 PMCID: PMC11176961 DOI: 10.1182/bloodadvances.2023010830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/28/2024] Open
Abstract
ABSTRACT Elevated MAPK and the JAK-STAT signaling play pivotal roles in the pathogenesis of chronic neutrophilic leukemia and atypical chronic myeloid leukemia. Although inhibitors targeting these pathways effectively suppress the diseases, they fall short in providing enduring remission, largely attributed to the cytostatic nature of these drugs. Even combinations of these drugs are ineffective in achieving sustained remission. Enhanced MAPK signaling besides promoting proliferation and survival triggers a proapoptotic response. Consequently, malignancies reliant on elevated MAPK signaling use MAPK feedback regulators to intricately modulate the signaling output, prioritizing proliferation and survival while dampening the apoptotic stimuli. Herein, we demonstrate that enhanced MAPK signaling in granulocyte colony-stimulating factor 3 receptor (CSF3R)-driven leukemia upregulates the expression of dual specificity phosphatase 1 (DUSP1) to suppress the apoptotic stimuli crucial for leukemogenesis. Consequently, genetic deletion of Dusp1 in mice conferred synthetic lethality to CSF3R-induced leukemia. Mechanistically, DUSP1 depletion in leukemic context causes activation of JNK1/2 that results in induced expression of BIM and P53 while suppressing the expression of BCL2 that selectively triggers apoptotic response in leukemic cells. Pharmacological inhibition of DUSP1 by BCI (a DUSP1 inhibitor) alone lacked antileukemic activity due to ERK1/2 rebound caused by off-target inhibition of DUSP6. Consequently, a combination of BCI with a MEK inhibitor successfully cured CSF3R-induced leukemia in a preclinical mouse model. Our findings underscore the pivotal role of DUSP1 in leukemic transformation driven by enhanced MAPK signaling and advocate for the development of a selective DUSP1 inhibitor for curative treatment outcomes.
Collapse
Affiliation(s)
- Meenu Kesarwani
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
| | - Zachary Kincaid
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
| | - Mohammad Azhar
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
| | - Mohammad Azam
- Division of Pathology, Cincinnati Children’s Hospital, Cincinnati, OH
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
29
|
Bjorklund GR, Rees KP, Balasubramanian K, Hewitt LT, Nishimura K, Newbern JM. Hyperactivation of MEK1 in cortical glutamatergic neurons results in projection axon deficits and aberrant motor learning. Dis Model Mech 2024; 17:dmm050570. [PMID: 38826084 PMCID: PMC11247507 DOI: 10.1242/dmm.050570] [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: 11/06/2023] [Accepted: 05/21/2024] [Indexed: 06/04/2024] Open
Abstract
Abnormal extracellular signal-regulated kinase 1/2 (ERK1/2, encoded by Mapk3 and Mapk1, respectively) signaling is linked to multiple neurodevelopmental diseases, especially the RASopathies, which typically exhibit ERK1/2 hyperactivation in neurons and non-neuronal cells. To better understand how excitatory neuron-autonomous ERK1/2 activity regulates forebrain development, we conditionally expressed a hyperactive MEK1 (MAP2K1) mutant, MEK1S217/221E, in cortical excitatory neurons of mice. MEK1S217/221E expression led to persistent hyperactivation of ERK1/2 in cortical axons, but not in soma/nuclei. We noted reduced axonal arborization in multiple target domains in mutant mice and reduced the levels of the activity-dependent protein ARC. These changes did not lead to deficits in voluntary locomotion or accelerating rotarod performance. However, skilled motor learning in a single-pellet retrieval task was significantly diminished in these MEK1S217/221E mutants. Restriction of MEK1S217/221E expression to layer V cortical neurons recapitulated axonal outgrowth deficits but did not affect motor learning. These results suggest that cortical excitatory neuron-autonomous hyperactivation of MEK1 is sufficient to drive deficits in axon outgrowth, which coincide with reduced ARC expression, and deficits in skilled motor learning. Our data indicate that neuron-autonomous decreases in long-range axonal outgrowth may be a key aspect of neuropathogenesis in RASopathies.
Collapse
Affiliation(s)
- George R. Bjorklund
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Katherina P. Rees
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | | | - Lauren T. Hewitt
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Kenji Nishimura
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Jason M. Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
30
|
Song G, Zhao F, Ni R, Deng B, Chen S, Hu R, Zheng J, Peng Y, Liu H, Luo Y, Zhou Z, Huang G, Shen W. Epithelial cells derived exosomal miR-203a-3p facilitates stromal inflammation of type IIIA chronic prostatitis/chronic pelvic pain syndrome by targeting DUSP5 and increasing MCP-1 generation. J Nanobiotechnology 2024; 22:236. [PMID: 38724995 PMCID: PMC11084011 DOI: 10.1186/s12951-024-02513-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
Increased proinflammatory cytokines and infiltration of inflammatory cells in the stroma are important pathological features of type IIIA chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS-A), and the interaction between stromal cells and other cells in the inflammatory microenvironment is closely related to the inflammatory process of CP/CPPS-A. However, the interaction between stromal and epithelial cells remains unclear. In this study, inflammatory prostate epithelial cells (PECs) released miR-203a-3p-rich exosomes and facilitated prostate stromal cells (PSCs) inflammation by upregulating MCP-1 expression. Mechanistically, DUSP5 was identified as a novel target gene of miR-203a-3p and regulated PSCs inflammation through the ERK1/2/MCP-1 signaling pathway. Meanwhile, the effect of exosomes derived from prostatic fluids of CP/CPPS-A patients was consistent with that of exosomes derived from inflammatory PECs. Importantly, we demonstrated that miR-203a-3p antagomirs-loaded exosomes derived from PECs targeted the prostate and alleviated prostatitis by inhibiting the DUSP5-ERK1/2 pathway. Collectively, our findings provide new insights into underlying the interaction between PECs and PSCs in CP/CPPS-A, providing a promising therapeutic strategy for CP/CPPS-A.
Collapse
Affiliation(s)
- Guojing Song
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Fuhan Zhao
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Rongrong Ni
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Bingqian Deng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Saipeng Chen
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Ruimin Hu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jun Zheng
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yiji Peng
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Heting Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yang Luo
- Department of Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400044, China
| | - Zhansong Zhou
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Gang Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Wenhao Shen
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
31
|
Ferruzo PYM, Boell VK, Russo LC, Oliveira CC, Forti FL. DUSP3 modulates IRES-dependent translation of mRNAs through dephosphorylation of the HNRNPC protein in cells under genotoxic stimulus. Biol Cell 2024; 116:e2300128. [PMID: 38538536 DOI: 10.1111/boc.202300128] [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: 12/22/2023] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 05/09/2024]
Abstract
BACKGROUND INFORMATION The dual-specificity phosphatase 3 (DUSP3) regulates cell cycle progression, proliferation, senescence, and DNA repair pathways under genotoxic stress. This phosphatase interacts with HNRNPC protein suggesting an involvement in the regulation of HNRNPC-ribonucleoprotein complex stability. In this work, we investigate the impact of DUSP3 depletion on functions of HNRNPC aiming to suggest new roles for this enzyme. RESULTS The DUSP3 knockdown results in the tyrosine hyperphosphorylation state of HNRNPC increasing its RNA binding ability. HNRNPC is present in the cytoplasm where it interacts with IRES trans-acting factors (ITAF) complex, which recruits the 40S ribosome on mRNA during protein synthesis, thus facilitating the translation of mRNAs containing IRES sequence in response to specific stimuli. In accordance with that, we found that DUSP3 is present in the 40S, monosomes and polysomes interacting with HNRNPC, just like other previously identified DUSP3 substrates/interacting partners such as PABP and NCL proteins. By downregulating DUSP3, Tyr-phosphorylated HNRNPC preferentially binds to IRES-containing mRNAs within ITAF complexes preferentially in synchronized or stressed cells, as evidenced by the higher levels of proteins such as c-MYC and XIAP, but not their mRNAs such as measured by qPCR. Under DUSP3 absence, this increased phosphorylated-HNRNPC/RNA interaction reduces HNRNPC-p53 binding in presence of RNAs releasing p53 for specialized cellular responses. Similarly, to HNRNPC, PABP physically interacts with DUSP3 in an RNA-dependent manner. CONCLUSIONS AND SIGNIFICANCE Overall, DUSP3 can modulate cellular responses to genotoxic stimuli at the translational level by maintaining the stability of HNRNPC-ITAF complexes and regulating the intensity and specificity of RNA interactions with RRM-domain proteins.
Collapse
Affiliation(s)
- Pault Y M Ferruzo
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Viktor K Boell
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Lilian C Russo
- Laboratory of Genome Instability, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Carla C Oliveira
- Laboratory of Post-transcriptional Control of Gene Expression, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Fabio L Forti
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
32
|
Deng Y, Deng X, Zhao J, Ning S, Gu A, Chen Q, Qu Y. Revealing the Complete Bispecific Phosphatase Genes (DUSPs) across the Genome and Investigating the Expression Patterns of GH_A11G3500 Resistance against Verticillium wilt. Int J Mol Sci 2024; 25:4500. [PMID: 38674085 PMCID: PMC11050305 DOI: 10.3390/ijms25084500] [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: 02/26/2024] [Revised: 04/07/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
DUSPs, a diverse group of protein phosphatases, play a pivotal role in orchestrating cellular growth and development through intricate signaling pathways. Notably, they actively participate in the MAPK pathway, which governs crucial aspects of plant physiology, including growth regulation, disease resistance, pest resistance, and stress response. DUSP is a key enzyme, and it is the enzyme that limits the rate of cell metabolism. At present, complete understanding of the DUSP gene family in cotton and its specific roles in resistance to Verticillium wilt (VW) remains elusive. To address this knowledge gap, we conducted a comprehensive identification and analysis of four key cotton species: Gossypium arboreum, Gossypium barbadense, Gossypium hirsutum, and Gossypium raimondii. The results revealed the identification of a total of 120 DUSP genes in the four cotton varieties, which were categorized into six subgroups and randomly distributed at both ends of 26 chromosomes, predominantly localized within the nucleus. Our analysis demonstrated that closely related DUSP genes exhibited similarities in terms of the conserved motif composition and gene structure. A promoter analysis performed on the GhDUSP gene promoter revealed the presence of several cis-acting elements, which are associated with abiotic and biotic stress responses, as well as hormone signaling. A tissue expression pattern analysis demonstrated significant variations in GhDUSP gene expression under different stress conditions, with roots exhibiting the highest levels, followed by stems and leaves. In terms of tissue-specific detection, petals, leaves, stems, stamens, and receptacles exhibited higher expression levels of the GhDUSP gene. The gene expression analysis results for GhDUSPs under stress suggest that DUSP genes may have a crucial role in the cotton response to stress in cotton. Through Virus-Induced Gene Silencing (VIGS) experiments, the silencing of the target gene significantly reduced the resistance efficiency of disease-resistant varieties against Verticillium wilt (VW). Consequently, we conclude that GH_A11G3500-mediated bispecific phosphorylated genes may serve as key regulators in the resistance of G. hirsutum to Verticillium wilt (VW). This study presents a comprehensive structure designed to provide an in-depth understanding of the potential biological functions of cotton, providing a strong foundation for further research into molecular breeding and resistance to plant pathogens.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yanying Qu
- College of Agronomy, Xinjiang Agricultural University, 311 Nongda East Road, Urumqi 830052, China; (Y.D.); (X.D.); (J.Z.); (S.N.); (A.G.); (Q.C.)
| |
Collapse
|
33
|
Zhu Y, Shi R, Lu W, Shi S, Chen Y. Framework nucleic acids as promising reactive oxygen species scavengers for anti-inflammatory therapy. NANOSCALE 2024; 16:7363-7377. [PMID: 38411498 DOI: 10.1039/d3nr05844a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Reactive oxygen species (ROS) are an array of derivatives of molecular oxygen that participate in multiple physiological processes under the control of redox homeostasis. However, under pathological conditions, the over-production of ROS often leads to oxidative stress and inflammatory reactions, indicating a potential therapeutic target. With the rapid development of nucleic acid nanotechnology, scientists have exploited various DNA nanostructures with remarkable biocompatibility, programmability, and structural stability. Among these novel organic nanomaterials, a group of skeleton-like framework nucleic acid (FNA) nanostructures attracts the most interest due to their outstanding self-assembly, cellular endocytosis, addressability, and functionality. Surprisingly, different FNAs manifest similarly satisfactory antioxidative and anti-inflammatory effects during their biomedical application process. First, they are intrinsically endowed with the ability to neutralize ROS due to their DNA nature. Therefore, they are extensively involved in the complicated inflammatory signaling network. Moreover, the outstanding editability of FNAs also allows for flexible modifications with nucleic acids, aptamers, peptides, antibodies, low-molecular-weight drugs, and so on, thus further strengthening the targeting and therapeutic ability. This review focuses on the ROS-scavenging potential of three representative FNAs, including tetrahedral framework nucleic acids (tFNAs), DNA origami, and DNA hydrogels, to summarize the recent advances in their anti-inflammatory therapy applications. Although FNAs exhibit great potential in treating inflammatory diseases as promising ROS scavengers, massive efforts still need to be made to overcome the emerging challenges in their clinical translation.
Collapse
Affiliation(s)
- Yujie Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Ruijianghan Shi
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Weitong Lu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Sirong Shi
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Yang Chen
- Department of Pediatric Surgery, Department of Liver Surgery & Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
34
|
Yokosawa T, Miyagawa S, Suzuki W, Nada Y, Hirata Y, Noguchi T, Matsuzawa A. The E3 Ubiquitin Protein Ligase LINCR Amplifies the TLR-Mediated Signals through Direct Degradation of MKP1. Cells 2024; 13:687. [PMID: 38667302 PMCID: PMC11048823 DOI: 10.3390/cells13080687] [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: 12/15/2023] [Revised: 04/13/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Toll-like receptors (TLRs) induce innate immune responses through activation of intracellular signaling pathways, such as MAP kinase and NF-κB signaling pathways, and play an important role in host defense against bacterial or viral infections. Meanwhile, excessive activation of TLR signaling leads to a variety of inflammatory disorders, including autoimmune diseases. TLR signaling is therefore strictly controlled to balance optimal immune response and inflammation. However, its balancing mechanisms are not fully understood. In this study, we identified the E3 ubiquitin ligase LINCR/ NEURL3 as a critical regulator of TLR signaling. In LINCR-deficient cells, the sustained activation of JNK and p38 MAPKs induced by the agonists for TLR3, TLR4, and TLR5, was clearly attenuated. Consistent with these observations, TLR-induced production of a series of inflammatory cytokines was significantly attenuated, suggesting that LINCR positively regulates innate immune responses by promoting the activation of JNK and p38. Interestingly, our further mechanistic study identified MAPK phosphatase-1 (MKP1), a negative regulator of MAP kinases, as a ubiquitination target of LINCR. Thus, our results demonstrate that TLRs fine-tune the activation of MAP kinase pathways by balancing LINCR (the positive regulator) and MKP1 (the negative regulator), which may contribute to the induction of optimal immune responses.
Collapse
Affiliation(s)
| | | | | | | | | | - Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| |
Collapse
|
35
|
Gallo MT, Dolci B, Fumagalli F, Brivio P, Calabrese F. Prenatal Fluoxetine Exposure Influences Glucocorticoid Receptor-Mediated Activity in the Prefrontal Cortex of Adolescent Rats Exposed to Acute Stress. ACS Chem Neurosci 2024; 15:1560-1569. [PMID: 38507566 DOI: 10.1021/acschemneuro.3c00856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024] Open
Abstract
Any deviation from the programmed processes of brain development may modify its formation and functions, thereby precipitating pathological conditions, which often become manifest in adulthood. Exposure to a challenge during crucial periods of vulnerability, such as adolescence, may reveal molecular changes preceding behavioral outcomes. Based on a previous study showing that prenatal fluoxetine (FLX) leads to the development of an anhedonic-like behavior in adult rats, we aimed to assess whether the same treatment regimen (i.e., fluoxetine during gestation; 15 mg/kg/day) influences the ability to respond to acute restraint stress (ARS) during adolescence. We subjected the rats to a battery of behavioral tests evaluating the development of various phenotypes (cognitive deficit, anhedonia, and anxiety). Furthermore, we carried out molecular analyses in the plasma and prefrontal cortex, a brain region involved in stress response, and whose functions are commonly altered in neuropsychiatric conditions. Our findings confirm that prenatal manipulation did not affect behavior in adolescent rats but impaired the capability to respond properly to ARS. Indeed, we observed changes in several molecular key players of the hypothalamic pituitary adrenal axis, particularly influencing genomic effects mediated by the glucocorticoid receptor. This study highlights that prenatal FLX exposure influences the ability of adolescent male rats to respond to an acute challenge, thereby altering the functionality of the hypothalamic-pituitary-adrenal axis, and indicates that the prenatal manipulation may prime the response to challenging events during this critical period of life.
Collapse
Affiliation(s)
- Maria Teresa Gallo
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan 20133, Italy
| | - Beatrice Dolci
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan 20133, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan 20133, Italy
| | - Paola Brivio
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan 20133, Italy
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan 20133, Italy
| |
Collapse
|
36
|
Bulle A, Liu P, Seehra K, Bansod S, Chen Y, Zahra K, Somani V, Khawar IA, Chen HP, Dodhiawala PB, Li L, Geng Y, Mo CK, Mahsl J, Ding L, Govindan R, Davies S, Mudd J, Hawkins WG, Fields RC, DeNardo DG, Knoerzer D, Held JM, Grierson PM, Wang-Gillam A, Ruzinova MB, Lim KH. Combined KRAS-MAPK pathway inhibitors and HER2-directed drug conjugate is efficacious in pancreatic cancer. Nat Commun 2024; 15:2503. [PMID: 38509064 PMCID: PMC10954758 DOI: 10.1038/s41467-024-46811-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 03/11/2024] [Indexed: 03/22/2024] Open
Abstract
Targeting the mitogen-activated protein kinase (MAPK) cascade in pancreatic ductal adenocarcinoma (PDAC) remains clinically unsuccessful. We aim to develop a MAPK inhibitor-based therapeutic combination with strong preclinical efficacy. Utilizing a reverse-phase protein array, we observe rapid phospho-activation of human epidermal growth factor receptor 2 (HER2) in PDAC cells upon pharmacological MAPK inhibition. Mechanistically, MAPK inhibitors lead to swift proteasomal degradation of dual-specificity phosphatase 6 (DUSP6). The carboxy terminus of HER2, containing a TEY motif also present in extracellular signal-regulated kinase 1/2 (ERK1/2), facilitates binding with DUSP6, enhancing its phosphatase activity to dephosphorylate HER2. In the presence of MAPK inhibitors, DUSP6 dissociates from the protective effect of the RING E3 ligase tripartite motif containing 21, resulting in its degradation. In PDAC patient-derived xenograft (PDX) models, combining ERK and HER inhibitors slows tumour growth and requires cytotoxic chemotherapy to achieve tumour regression. Alternatively, MAPK inhibitors with trastuzumab deruxtecan, an anti-HER2 antibody conjugated with cytotoxic chemotherapy, lead to sustained tumour regression in most tested PDXs without causing noticeable toxicity. Additionally, KRAS inhibitors also activate HER2, supporting testing the combination of KRAS inhibitors and trastuzumab deruxtecan in PDAC. This study identifies a rational and promising therapeutic combination for clinical testing in PDAC patients.
Collapse
Affiliation(s)
- Ashenafi Bulle
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Peng Liu
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kuljeet Seehra
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sapana Bansod
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yali Chen
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kiran Zahra
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Vikas Somani
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Iftikhar Ali Khawar
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hung-Po Chen
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Paarth B Dodhiawala
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lin Li
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yutong Geng
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chia-Kuei Mo
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jay Mahsl
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Li Ding
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ramaswamy Govindan
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sherri Davies
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jacqueline Mudd
- Section of Hepatobiliary Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - William G Hawkins
- Section of Hepatobiliary Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ryan C Fields
- Section of Hepatobiliary Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David G DeNardo
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | | | - Jason M Held
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Patrick M Grierson
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Andrea Wang-Gillam
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Marianna B Ruzinova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
37
|
Wang H, Irigoyen S, Liu J, Ramasamy M, Padilla C, Bedre R, Yang C, Thapa SP, Mulgaonkar N, Ancona V, He P, Coaker G, Fernando S, Mandadi KK. Inhibition of a conserved bacterial dual-specificity phosphatase confers plant tolerance to Candidatus Liberibacter spp. iScience 2024; 27:109232. [PMID: 38425843 PMCID: PMC10904284 DOI: 10.1016/j.isci.2024.109232] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/08/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024] Open
Abstract
"Candidatus Liberibacter spp." are insect-vectored, fastidious, and vascular-limited phytopathogens. They are the presumptive causal agents of potato zebra chip, tomato vein clearing, and the devastating citrus greening disease worldwide. There is an urgent need to develop new strategies to control them. In this study, we characterized a dual-specificity serine/tyrosine phosphatase (STP) that is well conserved among thirty-three geographically diverse "Candidatus Liberibacter spp." and strains that infect multiple Solanaceaea and citrus spp. The STP is expressed in infected plant tissues, localized at the plant cytosol and plasma membrane, and interferes with plant cell death responses. We employed an in silico target-based molecular modeling and ligand screen to identify two small molecules with high binding affinity to STP. Efficacy studies demonstrated that the two molecules can inhibit "Candidatus Liberibacter spp." but not unrelated pathogens and confer plant disease tolerance. The inhibitors and strategies are promising means to control "Candidatus Liberibacter spp."
Collapse
Affiliation(s)
- Haoqi Wang
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX, USA
| | - Sonia Irigoyen
- Texas A&M AgriLife Research & Extension Center, Texas A&M University System, 2415 E. Highway 83, Weslaco TX 78596, USA
| | - Jiaxing Liu
- Texas A&M AgriLife Research & Extension Center, Texas A&M University System, 2415 E. Highway 83, Weslaco TX 78596, USA
| | - Manikandan Ramasamy
- Texas A&M AgriLife Research & Extension Center, Texas A&M University System, 2415 E. Highway 83, Weslaco TX 78596, USA
| | - Carmen Padilla
- Texas A&M AgriLife Research & Extension Center, Texas A&M University System, 2415 E. Highway 83, Weslaco TX 78596, USA
| | - Renesh Bedre
- Texas A&M AgriLife Research & Extension Center, Texas A&M University System, 2415 E. Highway 83, Weslaco TX 78596, USA
| | - Chuanyu Yang
- Department of Agriculture, Agribusiness, and Environmental Sciences, Texas A&M University-Kingsville, Citrus Center, Weslaco, TX, USA
| | - Shree P. Thapa
- Department of Plant Pathology, University of California, Davis, Davis, CA, USA
| | - Nirmitee Mulgaonkar
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX, USA
| | - Veronica Ancona
- Department of Agriculture, Agribusiness, and Environmental Sciences, Texas A&M University-Kingsville, Citrus Center, Weslaco, TX, USA
| | - Ping He
- Department of Biochemistry and Biophysics, Texas A&M University, 2128 TAMU, College Station, TX, USA
| | - Gitta Coaker
- Department of Plant Pathology, University of California, Davis, Davis, CA, USA
| | - Sandun Fernando
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX, USA
| | - Kranthi K. Mandadi
- Texas A&M AgriLife Research & Extension Center, Texas A&M University System, 2415 E. Highway 83, Weslaco TX 78596, USA
- Department of Plant Pathology and Microbiology, Texas A&M University, 2132 TAMU, College Station, TX, USA
- Institute for Advancing Health Through Agriculture, Texas A&M AgriLife, College Station, TX, USA
| |
Collapse
|
38
|
Chen L, Su H, Tao Z, Liang C, Liu Z, Dong Y, Zheng P, Liu Y. DUSP22 Ameliorates Endothelial-to-Mesenchymal Transition in HUVECs through Smad2/3 and MAPK Signaling Pathways. Cardiovasc Ther 2024; 2024:5583961. [PMID: 38495810 PMCID: PMC10942825 DOI: 10.1155/2024/5583961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 03/19/2024] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT) is the process by which endothelial cells lose their endothelial properties and acquire mesenchymal characteristics. Dual-specific protein phosphatase 22 (DUSP22) inactivates various protein kinases and transcription factors by dephosphorylating serine/threonine residues: hence, it plays a key role in many diseases. The aim of this study was to explore the functional role of DUSP22 in EndMT. In the transforming growth factor-β-induced EndMT model in human umbilical vein endothelial cells (HUVECs), we observed a downregulation of DUSP22 expression. This DUSP22 deficiency could aggravate EndMT. Conversely, the overexpression of DUSP22 could ameliorate EndMT. We used signaling pathway inhibitors to verify our results and found that DUSP22 could regulate EndMT through the smad2/3 and the mitogen-activated protein kinase (MAPK) signaling pathways. In summary, DUSP22 ameliorates EndMT in HUVECs in vitro through the smad2/3 and MAPK signaling pathways.
Collapse
Affiliation(s)
- Lu Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongyu Su
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zekai Tao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cui Liang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongzhao Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiming Dong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peipei Zheng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuan Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
39
|
Hao SH, Ma XD, Xu L, Xie JD, Feng ZH, Chen JW, Chen RX, Wang FW, Tang YH, Xie D, Cai MY. Dual specific phosphatase 4 suppresses ferroptosis and enhances sorafenib resistance in hepatocellular carcinoma. Drug Resist Updat 2024; 73:101052. [PMID: 38262246 DOI: 10.1016/j.drup.2024.101052] [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: 09/23/2023] [Revised: 12/26/2023] [Accepted: 01/06/2024] [Indexed: 01/25/2024]
Abstract
AIMS This investigation aims to elucidate the mechanism underlying sorafenib-induced ferroptosis in hepatocellular carcinoma (HCC). METHODS The role of dual specificity phosphatase 4 (DUSP4) in sorafenib-treated HCC was investigated using comprehensive assessments both in vitro and in vivo, including Western blotting, qRT-PCR, cell viability assay, lipid reactive oxygen species (ROS) assay, immunohistochemistry, and xenograft tumor mouse model. Additionally, label-free quantitative proteomics was employed to identify potential proteins associated with DUSP4. RESULTS Our study revealed that suppression of DUSP4 expression heightens the susceptibility of HCC cells to ferroptosis inducers, specifically sorafenib and erastin, in both in vitro and in vivo settings. Furthermore, we identified DUSP4-mediated regulation of key ferroptosis-related markers, such as ferritin light chain (FTL) and ferritin heavy chain 1 (FTH1). Notably, label-free quantitative proteomics unveiled the phosphorylation of threonine residue T148 on YTH Domain Containing 1 (YTHDC1) by DUSP4. Further investigations unraveled that YTHDC1, functioning as an mRNA nuclear export regulator, is a direct target of DUSP4, orchestrating the subcellular localization of FTL and FTH1 mRNAs. Significantly, our study highlights a strong correlation between elevated DUSP4 expression and sorafenib resistance in HCC. CONCLUSIONS Our findings introduce DUSP4 as a negative regulator of sorafenib-induced ferroptosis. This discovery opens new avenues for the development of ferroptosis-based therapeutic strategies tailored for HCC treatment.
Collapse
Affiliation(s)
- Shi-Hui Hao
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xiao-Dan Ma
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 510060 Guangzhou, China
| | - Li Xu
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jing-Dun Xie
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Zi-Hao Feng
- Department of Urology, the First Affiliated Hospital, Sun Yat-sen University, 510060 Guangzhou, China
| | - Jie-Wei Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 510060 Guangzhou, China; Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ri-Xin Chen
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Department of Thoracic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Feng-Wei Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 510060 Guangzhou, China
| | - Yu-Hao Tang
- Department of Liver Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 510060 Guangzhou, China; Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Mu-Yan Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 510060 Guangzhou, China; Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
40
|
Elahimanesh M, Shokri N, Mahdinia E, Mohammadi P, Parvaz N, Najafi M. Differential gene expression patterns in ST-elevation Myocardial Infarction and Non-ST-elevation Myocardial Infarction. Sci Rep 2024; 14:3424. [PMID: 38341440 PMCID: PMC10858964 DOI: 10.1038/s41598-024-54086-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/08/2024] [Indexed: 02/12/2024] Open
Abstract
The ST-elevation Myocardial Infarction (STEMI) and Non-ST-elevation Myocardial Infarction (NSTEMI) might occur because of coronary artery stenosis. The gene biomarkers apply to the clinical diagnosis and therapeutic decisions in Myocardial Infarction. The aim of this study was to introduce, enrich and estimate timely the blood gene profiles based on the high-throughput data for the molecular distinction of STEMI and NSTEMI. The text mining data (50 genes) annotated with DisGeNET data (144 genes) were merged with the GEO gene expression data (5 datasets) using R software. Then, the STEMI and NSTEMI networks were primarily created using the STRING server, and improved using the Cytoscape software. The high-score genes were enriched using the KEGG signaling pathways and Gene Ontology (GO). Furthermore, the genes were categorized to determine the NSTEMI and STEMI gene profiles. The time cut-off points were identified statistically by monitoring the gene profiles up to 30 days after Myocardial Infarction (MI). The gene heatmaps were clearly created for the STEMI (high-fold genes 69, low-fold genes 45) and NSTEMI (high-fold genes 68, low-fold genes 36). The STEMI and NSTEMI networks suggested the high-score gene profiles. Furthermore, the gene enrichment suggested the different biological conditions for STEMI and NSTEMI. The time cut-off points for the NSTEMI (4 genes) and STEMI (13 genes) gene profiles were established up to three days after Myocardial Infarction. The study showed the different pathophysiologic conditions for STEMI and NSTEMI. Furthermore, the high-score gene profiles are suggested to measure up to 3 days after MI to distinguish the STEMI and NSTEMI.
Collapse
Affiliation(s)
- Mohammad Elahimanesh
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Shokri
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Elmira Mahdinia
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Payam Mohammadi
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Najmeh Parvaz
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
41
|
Lin P, Niimi H, Hirota T, Ohsugi Y, Shimohira T, Toyoshima K, Katagiri S, Iwata T, Aoki A. Effects of low-level Er:YAG laser irradiation on proliferation and gene expression in primary gingival fibroblasts isolated from mouse maxilla. JOURNAL OF BIOPHOTONICS 2024; 17:e202300166. [PMID: 37975254 DOI: 10.1002/jbio.202300166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 10/06/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023]
Abstract
We investigated the effects of low-level Er:YAG laser irradiation on proliferation and alternations in early gene expression of gingival fibroblasts. Mice primary gingival fibroblasts were irradiated with an Er:YAG laser (1.8, 3.9, and 5.8 J/cm2 ). Irradiation at 3.9 J/cm2 promoted cell proliferation without significant changes in lactate dehydrogenase or Hspa1a expression. Three hours after irradiation at 3.9 J/cm2 , the Fn1 expression level was significantly increased. RNA-seq identified 15 differentially expressed genes between irradiated and non-irradiated cells, some of which belonged to immediate early genes (IEGs). Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated MAPK pathway enhancement, and gene set enrichment analysis showed enrichment in the TGF-β signaling gene set. Enhanced proliferation via laser irradiation disappeared upon inhibition of Dusp4, Dusp5, and Tgfr1 expression. Low-level Er:YAG laser irradiation, especially at 3.9 J/cm2 without a major temperature elevation, enhanced fibroblast proliferation, via TGF-β and the MAPK signaling pathway following IEG expression.
Collapse
Affiliation(s)
- Peiya Lin
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiromi Niimi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tomomitsu Hirota
- Division of Molecular Genetics, Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Yujin Ohsugi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tsuyoshi Shimohira
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Keita Toyoshima
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Sayaka Katagiri
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Akira Aoki
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
42
|
Cosgrove BD, Bounds LR, Taylor CK, Su AL, Rizzo AJ, Barrera A, Crawford GE, Hoffman BD, Gersbach CA. Mechanosensitive genomic enhancers potentiate the cellular response to matrix stiffness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.574997. [PMID: 38260455 PMCID: PMC10802421 DOI: 10.1101/2024.01.10.574997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Epigenetic control of cellular transcription and phenotype is influenced by changes in the cellular microenvironment, yet how mechanical cues from these microenvironments precisely influence epigenetic state to regulate transcription remains largely unmapped. Here, we combine genome-wide epigenome profiling, epigenome editing, and phenotypic and single-cell RNA-seq CRISPR screening to identify a new class of genomic enhancers that responds to the mechanical microenvironment. These 'mechanoenhancers' could be active on either soft or stiff extracellular matrix contexts, and regulated transcription to influence critical cell functions including apoptosis, mechanotransduction, proliferation, and migration. Epigenetic editing of mechanoenhancers on rigid materials tuned gene expression to levels observed on softer materials, thereby reprogramming the cellular response to the mechanical microenvironment. These editing approaches may enable the precise alteration of mechanically-driven disease states.
Collapse
Affiliation(s)
- Brian D. Cosgrove
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Lexi R. Bounds
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Carson Key Taylor
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Alan L. Su
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Anthony J. Rizzo
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Alejandro Barrera
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
- Department of Biostatistics and Bioinformatics, Duke University; Durham, NC 27708, USA
| | - Gregory E. Crawford
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
- Department of Pediatrics, Duke University Medical Center; Durham, NC 27708, USA
| | - Brenton D. Hoffman
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Department of Cell Biology, Duke University; Durham, NC 27708, USA
| | - Charles A. Gersbach
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
- Department of Cell Biology, Duke University; Durham, NC 27708, USA
- Department of Surgery, Duke University Medical Center; Durham, NC 27708, USA
| |
Collapse
|
43
|
Li J, Zhai Y, Tang M. Integrative function of histone deacetylase 3 in inflammation. Mol Biol Rep 2024; 51:83. [PMID: 38183491 DOI: 10.1007/s11033-023-09077-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/11/2023] [Indexed: 01/08/2024]
Abstract
Inflammation is a complex biological response triggered when an organism encounters internal or external stimuli. These triggers activate various signaling pathways, leading to the release of numerous inflammatory mediators aimed at the affected tissue. Ensuring precision and avoiding the excessive activation, the inflammatory process is subject to tight regulation. Histone deacetylase 3 (HDAC3), a member of class I HDACs family, stands out for its significant role in modulating various inflammatory signaling, including Nuclear Factor kappa B (NF-κB) signaling, Mitogen-activated protein kinase (MAPK) signaling and Janus kinase/signal transduction and activator of transcription (JAK-STAT) signaling. In this review, we illuminate the intricate molecular mechanisms of HDAC3 across these inflammatory pathways. We emphasize its importance in orchestrating a balanced inflammatory response and highlight its promising potential as a therapeutic target.
Collapse
Affiliation(s)
- Junjie Li
- Institute of Biochemistry and Molecular Biology, Hengyang College of Medicine, University of South China, Changsheng West Road 28, Hengyang, 421001, China
| | - Yiyuan Zhai
- Institute of Biochemistry and Molecular Biology, Hengyang College of Medicine, University of South China, Changsheng West Road 28, Hengyang, 421001, China
| | - Min Tang
- Institute of Biochemistry and Molecular Biology, Hengyang College of Medicine, University of South China, Changsheng West Road 28, Hengyang, 421001, China.
| |
Collapse
|
44
|
Mandal J, Yu ZC, Shih IM, Wang TL. ARID1A loss activates MAPK signaling via DUSP4 downregulation. J Biomed Sci 2023; 30:94. [PMID: 38071325 PMCID: PMC10709884 DOI: 10.1186/s12929-023-00985-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/12/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND ARID1A, a tumor suppressor gene encoding BAF250, a protein participating in chromatin remodeling, is frequently mutated in endometrium-related malignancies, including ovarian or uterine clear cell carcinoma (CCC) and endometrioid carcinoma (EMCA). However, how ARID1A mutations alter downstream signaling to promote tumor development is yet to be established. METHODS We used RNA-sequencing (RNA-seq) to explore transcriptomic changes in isogenic human endometrial epithelial cells after deleting ARID1A. Chromatin immunoprecipitation sequencing (ChIP-seq) was employed to assess the active or repressive histone marks on DUSP4 promoter and regulatory regions. We validated our findings using genetically engineered murine endometroid carcinoma models, human endometroid carcinoma tissues, and in silico approaches. RESULTS RNA-seq revealed the downregulation of the MAPK phosphatase dual-specificity phosphatase 4 (DUSP4) in ARID1A-deficient cells. ChIP-seq demonstrated decreased histone acetylation marks (H3K27Ac, H3K9Ac) on DUSP4 regulatory regions as one of the causes for DUSP4 downregulation in ARID1A-deficient cells. Ectopic DUSP4 expression decreased cell proliferation, and pharmacologically inhibiting the MAPK pathway significantly mitigated tumor formation in vivo. CONCLUSIONS Our findings suggest that ARID1A protein transcriptionally modulates DUSP4 expression by remodeling chromatin, subsequently inactivating the MAPK pathway, leading to tumor suppression. The ARID1A-DUSP4-MAPK axis may be further considered for developing targeted therapies against ARID1A-mutated cancers.
Collapse
Affiliation(s)
- Jayaprakash Mandal
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zheng-Cheng Yu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ie-Ming Shih
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Tian-Li Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
45
|
Shaikh N, Waterhölter A, Gnirck AC, Becker M, Adamiak V, Henneken L, Wunderlich M, Hartmann W, Linnemann L, Huber TB, Krebs CF, Panzer U, Locksley RM, Wilhelm C, Breloer M, Turner JE. Retinoic acid drives intestine-specific adaptation of effector ILC2s originating from distant sites. J Exp Med 2023; 220:e20221015. [PMID: 37773047 PMCID: PMC10541314 DOI: 10.1084/jem.20221015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/26/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023] Open
Abstract
Adaptation of immune cells to tissue-specific microenvironments is a crucial process in homeostasis and inflammation. Here, we show that murine effector type 2 innate lymphoid cells (ILC2s) from various organs are equally effective in repopulating ILC2 niches in other anatomical locations where they adapt tissue-specific phenotypes of target organs. Single-cell transcriptomics of ILC2 populations revealed upregulation of retinoic acid (RA) signaling in ILC2s during adaptation to the small intestinal microenvironment, and RA signaling mediated reprogramming of kidney effector ILC2s toward the small intestinal phenotype in vitro and in vivo. Inhibition of intestinal ILC2 adaptation by blocking RA signaling impaired worm expulsion during Strongyloides ratti infection, indicating functional importance of ILC2 tissue imprinting. In conclusion, this study highlights that effector ILC2s retain the ability to adapt to changing tissue-specific microenvironments, enabling them to exert tissue-specific functions, such as promoting control of intestinal helminth infections.
Collapse
Affiliation(s)
- Nikhat Shaikh
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alex Waterhölter
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ann-Christin Gnirck
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martina Becker
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Virginia Adamiak
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lena Henneken
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Wunderlich
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wiebke Hartmann
- Helminth Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Lara Linnemann
- Helminth Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian F. Krebs
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Division of Translational Immunology, III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Division of Translational Immunology, III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Richard M. Locksley
- Department of Medicine, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Christoph Wilhelm
- Unit for Immunopathology, Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Minka Breloer
- Helminth Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jan-Eric Turner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
46
|
Ko E, Yoon T, Lee Y, Kim J, Park YB. ADSC secretome constrains NK cell activity by attenuating IL-2-mediated JAK-STAT and AKT signaling pathway via upregulation of CIS and DUSP4. Stem Cell Res Ther 2023; 14:329. [PMID: 37964351 PMCID: PMC10648656 DOI: 10.1186/s13287-023-03516-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: 05/01/2023] [Accepted: 09/25/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have immunomodulatory properties and therapeutic effects on autoimmune diseases through their secreted factors, referred to as the secretome. However, the specific key factors of the MSC secretome and their mechanisms of action in immune cells have not been fully determined. Most in vitro experiments are being performed using immune cells, but experiments using natural killer (NK) cells have been neglected, and a few studies using NK cells have shown discrepancies in results. NK cells are crucial elements of the immune system, and adjustment of their activity is essential for controlling various pathological conditions. The aim of this study was to elucidate the role of the adipose tissue-derived stem cell (ADSC) secretome on NK cell activity. METHODS To obtain the ADSC secretome, we cultured ADSCs in medium and concentrated the culture medium using tangential flow filtration (TFF) capsules. We assessed NK cell viability and proliferation using CCK-8 and CFSE assays, respectively. We analyzed the effects of the ADSC secretome on NK cell activity and pathway-related proteins using a combination of flow cytometry, ELISA, cytotoxicity assay, CD107a assay, western blotting, and quantitative real-time PCR. To identify the composition of the ADSC secretome, we performed LC-MS/MS profiling and bioinformatics analysis. To elucidate the molecular mechanisms involved, we used mRNA sequencing to profile the transcriptional expression of human blood NK cells. RESULTS The ADSC secretome was found to restrict IL-2-mediated effector function of NK cells while maintaining proliferative potency. This effect was achieved through the upregulation of the inhibitory receptor CD96, as well as downregulation of activating receptors and IL-2 receptor subunits IL-2Rα and IL-2Rγ. These changes were associated with attenuated JAK-STAT and AKT pathways in NK cells, which were achieved through the upregulation of cytokine-inducible SH2-containing protein (CIS, encoded by Cish) and dual specificity protein phosphatase 4 (DUSP4). Furthermore, proteomic analysis revealed twelve novel candidates associated with the immunomodulatory effects of MSCs. CONCLUSIONS Our findings reveal a detailed cellular outcome and regulatory mechanism of NK cell activity by the ADSC secretome and suggest a therapeutic tool for treating NK-mediated inflammatory and autoimmune diseases using the MSC secretome.
Collapse
Affiliation(s)
- Eunhee Ko
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Taejun Yoon
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yoojin Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jongsun Kim
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
47
|
Ma C, Liu M, Feng W, Rao H, Zhang W, Liu C, Xu Y, Wang Z, Teng Y, Yang X, Ni L, Xu J, Gao W, Lu B, Li L. Loss of SETD2 aggravates colorectal cancer progression caused by SMAD4 deletion through the RAS/ERK signalling pathway. Clin Transl Med 2023; 13:e1475. [PMID: 37962020 PMCID: PMC10644329 DOI: 10.1002/ctm2.1475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGOUND Colorectal cancer (CRC) is a complex, multistep disease that arises from the interplay genetic mutations and epigenetic alterations. The histone H3K36 trimethyltransferase SET domain-containing 2 (SETD2), as an epigenetic signalling molecule, has a 5% mutation rate in CRC. SETD2 expression is decreased in the development of human CRC and mice treated with Azoxymethane /Dextran sodium sulfate (AOM/DSS). Loss of SETD2 promoted CRC development. SMAD Family member 4 (SMAD4) has a 14% mutation rate in CRC, and SMAD4 ablation leads to CRC. The co-mutation of SETD2 and SMAD4 predicted advanced CRC. However, little is known on the potential synergistic effect of SETD2 and SMAD4. METHODS CRC tissues from mice and SW620 cells were used as research subjects. Clinical databases of CRC patients were analyzed to investigate the association between SETD2 and SMAD4. SETD2 and SMAD4 double-knockout mice were established to further investigate the role of SETD2 in SMAD4-deficient CRC. The intestinal epithelial cells (IECs) were isolated for RNA sequencing and chromatin immunoprecipitation sequencing (ChIP-seq) to explore the mechanism and the key molecules resulting in CRC. Molecular and cellular experiments were conducted to analyze the role of SETD2 in SMAD4-deficient CRC. Finally, rescue experiments were performed to confirm the molecular mechanism of SETD2 in the development of SMAD4-dificient CRC. RESULTS The deletion of SETD2 promotes the malignant progression of SMAD4-deficient CRC. Smad4Vil-KO ; Setd2Vil-KO mice developed a more severe CRC phenotype after AOM/DSS induction, with a larger tumour size and a more vigorous epithelial proliferation rate. Further mechanistic findings revealed that the loss of SETD2 resulted in the down-regulation of DUSP7, which is involved in the inhibition of the RAS/ERK signalling pathway. Finally, the ERK1/2 inhibitor SCH772984 significantly attenuated the progression of CRC in Smad4Vil-KO ;Setd2Vil-KO mice, and overexpression of DUSP7 significantly inhibited the proliferation rates of SETD2KO ; SMAD4KO SW620 cells. CONCLUSIONS Our results demonstrated that SETD2 inhibits the RAS/ERK signaling pathway by facilitating the transcription of DUSP7 in SMAD4-deficient CRC, which could provide a potential therapeutic target for the treatment of advanced CRC.
Collapse
Affiliation(s)
- Chunxiao Ma
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research Institute, Shanghai Jiao Tong UniversityShanghaiChina
| | - Min Liu
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research Institute, Shanghai Jiao Tong UniversityShanghaiChina
| | - Wenxin Feng
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research Institute, Shanghai Jiao Tong UniversityShanghaiChina
| | - Hanyu Rao
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research Institute, Shanghai Jiao Tong UniversityShanghaiChina
| | - Wei Zhang
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research Institute, Shanghai Jiao Tong UniversityShanghaiChina
| | - Changwei Liu
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research Institute, Shanghai Jiao Tong UniversityShanghaiChina
| | - Yue Xu
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research Institute, Shanghai Jiao Tong UniversityShanghaiChina
| | - Ziyi Wang
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research Institute, Shanghai Jiao Tong UniversityShanghaiChina
| | - Yan Teng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein SciencesBeijing Institute of LifeomicsBeijingChina
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein SciencesBeijing Institute of LifeomicsBeijingChina
| | - Li Ni
- Department of NursingShanghai East Hospital, Tongji UniversityShanghaiChina
| | - Jin Xu
- School of Biomedical Engineering and Med‐X Research Institute, Shanghai Jiao Tong UniversityShanghaiChina
| | - Wei‐Qiang Gao
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research Institute, Shanghai Jiao Tong UniversityShanghaiChina
| | - Bing Lu
- Department of General Surgery, Department of Colorectal Surgery, Shanghai East HospitalSchool of Medicine, Tongji UniversityShanghaiChina
| | - Li Li
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiChina
- School of Biomedical Engineering and Med‐X Research Institute, Shanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
48
|
Kobayashi K, Iwasa K, Azuma-Suzuki R, Kawauchi T, Nabeshima YI. Feto-maternal cholesterol transport regulated by β-Klotho-FGF15 axis is essential for fetal growth. Life Sci Alliance 2023; 6:e202301916. [PMID: 37541847 PMCID: PMC10403640 DOI: 10.26508/lsa.202301916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/06/2023] Open
Abstract
β-Klotho (β-KL) is indispensable to regulate lipid, glucose, and energy metabolism in adult animals. β-KL is highly expressed in the yolk sac, but its role in the developmental stages has not been established. We hypothesized that β-KL is required for metabolic regulation in the embryo and aimed to clarify the role of β-KL during development. Here, we show that β-KL regulates feto-maternal cholesterol transport through the yolk sac by mediating FGF 15 signaling, and also that impairment of the β-KL-FGF15 axis causes fetal growth restriction (FGR). Embryos of β- kl knockout (β-kl-/-) mice were morphologically normal but exhibited FGR before placental maturation. The body weight of β-kl-/- mice remained lower after birth. β-KL deletion reduced cholesterol supply from the maternal blood and led to lipid shortage in the embryos. These phenotypes were similar to those of embryos lacking FGF15, indicating that β-KL-FGF15 axis is essential for growth and lipid regulation in the embryonic stages. Our findings suggest that lipid abnormalities in early gestation provoke FGR, leading to reduced body size in later life.
Collapse
Affiliation(s)
- Kanako Kobayashi
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Kazuko Iwasa
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Rika Azuma-Suzuki
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Takeshi Kawauchi
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Adaptive and Maladaptive Responses in Health and Disease, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Yo-Ichi Nabeshima
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| |
Collapse
|
49
|
Lee GH, Min CW, Jang JW, Wang Y, Jeon JS, Gupta R, Kim ST. Analysis of post-translational modification dynamics unveiled novel insights into Rice responses to MSP1. J Proteomics 2023; 287:104970. [PMID: 37467888 DOI: 10.1016/j.jprot.2023.104970] [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: 05/03/2023] [Revised: 07/04/2023] [Accepted: 07/09/2023] [Indexed: 07/21/2023]
Abstract
Magnaporthe oryzae snodprot1 homologous protein (MSP1) is known to function as a pathogen-associated molecular pattern (PAMP) and trigger PAMP-triggered immunity (PTI) in rice including induction of programmed cell death and expression of defense-related genes. The involvement of several post-translational modifications (PTMs) in the regulation of plant immune response, especially PTI, is well established, however, the information on the regulatory roles of these PTMs in response to MSP1-induced signaling is currently elusive. Here, we report the phosphoproteome, ubiquitinome, and acetylproteome to investigate the MSP1-induced PTMs alterations in MSP1 overexpressed and wild-type rice. Our analysis identified a total of 4666 PTMs-modified sites in rice leaves including 4292 phosphosites, 189 ubiquitin sites, and 185 acetylation sites. Among these, the PTM status of 437 phosphorylated, 53 ubiquitinated, and 68 acetylated peptides was significantly changed by MSP1. Functional annotation of MSP1 modulated peptides by MapMan analysis revealed that these were majorly associated with cellular immune responses including signaling, transcription factors, DNA and RNA regulation, and protein metabolism, among others. Taken together, our study provides novel insights into post-translational mediated regulation of rice proteins in response to M. oryzae secreted PAMP which help in understanding the molecular mechanism of MSP1-induced signaling in rice in greater detail. SIGNIFICANCE: The research investigates the effect of overexpression of MSP1 protein in rice leaves on the phosphoproteome, acetylome, and ubiquitinome. The study found that MSP1 is involved in rice protein phosphorylation, particularly in signaling pathways, and identified a key component, PTAC16, in MSP1-induced signaling. The analysis also revealed MSP1's role in protein degradation and modification by inducing ubiquitination of the target rice proteins. The research identified potential kinases involved in the phosphorylation of rice proteins, including casein kinase II, 14-3-3 domain binding motif, β-adrenergic receptor kinase, ERK1,2 kinase substrate motif, and casein kinase I motifs. Overall, the findings provide insights into the molecular mechanisms underlying of MSP1 induced signaling in rice which may have implications for improving crop yield and quality.
Collapse
Affiliation(s)
- Gi Hyun Lee
- Department of Plant Bioscience, Pusan National University, Miryang 50463, South Korea
| | - Cheol Woo Min
- Department of Plant Bioscience, Pusan National University, Miryang 50463, South Korea
| | - Jeong Woo Jang
- Department of Plant Bioscience, Pusan National University, Miryang 50463, South Korea
| | - Yiming Wang
- Key Laboratory of Integrated Management of Crop Disease and Pests, Ministry of Education, Department of Plant Pathology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Jong-Seong Jeon
- Graduate School of Biotechnology and Crop Biotech Institute, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Ravi Gupta
- College of General Education, Kookmin University, Seoul 02707, South Korea.
| | - Sun Tae Kim
- Department of Plant Bioscience, Pusan National University, Miryang 50463, South Korea.
| |
Collapse
|
50
|
Shi SX, Xiu Y, Li Y, Yuan M, Shi K, Liu Q, Wang X, Jin WN. CD4 + T cells aggravate hemorrhagic brain injury. SCIENCE ADVANCES 2023; 9:eabq0712. [PMID: 37285421 DOI: 10.1126/sciadv.abq0712] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/01/2023] [Indexed: 06/09/2023]
Abstract
Leukocyte infiltration accelerates brain injury following intracerebral hemorrhage (ICH). Yet, the involvement of T lymphocytes in this process has not been fully elucidated. Here, we report that CD4+ T cells accumulate in the perihematomal regions in the brains of patients with ICH and ICH mouse models. T cells activation in the ICH brain is concurrent with the course of perihematomal edema (PHE) development, and depletion of CD4+ T cells reduced PHE volumes and improved neurological deficits in ICH mice. Single-cell transcriptomic analysis revealed that brain-infiltrating T cells exhibited enhanced proinflammatory and proapoptotic signatures. Consequently, CD4+ T cells disrupt the blood-brain barrier integrity and promote PHE progression through interleukin-17 release; furthermore, the TRAIL-expressing CD4+ T cells engage DR5 to trigger endothelial death. Recognition of T cell contribution to ICH-induced neural injury is instrumental for designing immunomodulatory therapies for this dreadful disease.
Collapse
Affiliation(s)
- Samuel X Shi
- Clinical Neuroscience Research Center (CNRC), Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Yuwen Xiu
- Clinical Neuroscience Research Center (CNRC), Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA
- Center for Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Li
- Center for Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Meng Yuan
- Center for Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kaibin Shi
- Center for Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qiang Liu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoying Wang
- Clinical Neuroscience Research Center (CNRC), Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Wei-Na Jin
- Center for Neurological Diseases, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|