1
|
Senatore E, Avolio R, Rinaldi L, Chiuso F, Oliva MA, D'Ambrosio C, Bianco AG, Dalla E, Pagnotta SM, Flammia R, Ambrosino C, Memoli D, Turacchio G, Mimoune SI, Toiron Y, Audebert S, Camoin L, Lignitto L, Scaloni A, Arcella A, Feliciello A. Praja2 controls P-body assembly and translation in glioblastoma by non-proteolytic ubiquitylation of DDX6. EMBO Rep 2025; 26:2347-2377. [PMID: 40148504 PMCID: PMC12069581 DOI: 10.1038/s44319-025-00425-5] [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: 10/08/2024] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Glioblastoma multiforme (GBM) is the most lethal form of malignant brain tumor in adults. Dysregulation of protein synthesis contributes to cancer cell plasticity, driving GBM cell heterogeneity, metastatic behavior, and drug resistance. Understanding the complex network and signaling pathways governing protein translation, is therefore an important goal for GBM treatment. Here we identify a novel signaling network centered on the E3 ubiquitin ligase praja2 that controls protein translation in GBM. Praja2 forms a multimeric complex with the RNA helicase DDX6, which inhibits translation of target RNAs within processing bodies (P-bodies). Stimulation of cAMP signaling through activation of G-protein-coupled receptors induces P-body assembly through praja2-mediated non-proteolytic polyubiquitylation of DDX6. Genetic inactivation of praja2 reshapes DDX6/mRNA complexes and translating polysomes and promotes cellular senescence and GBM growth arrest. Expression of an ubiquitylation-defective DDX6 mutant suppresses the assembly of P-bodies and sustains GBM growth. Taken together, our findings identify a cAMP-driven network that controls translation in P-bodies and GBM growth.
Collapse
Affiliation(s)
- Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Rosario Avolio
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | | | - Chiara D'Ambrosio
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici, Naples, Italy
| | - Antonio Giuseppe Bianco
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Emiliano Dalla
- Department of Medicine, University of Udine, Udine, Italy
| | | | - Raffaella Flammia
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Concetta Ambrosino
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Domenico Memoli
- Department of Medicine, Surgery and Dentistry SMS, University of Salerno, Salerno, Italy
| | - Gabriele Turacchio
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Sonia Ines Mimoune
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Yves Toiron
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Stephane Audebert
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Luc Camoin
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Luca Lignitto
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Andrea Scaloni
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici, Naples, Italy
| | | | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy.
| |
Collapse
|
2
|
Chen Z, Peng C, Jin C, Wang Y, Wang T, Yang P, Peng W, Sun Q, Xu H, Nie H, Wang X, Tang J, Sun Y, Feng Y. PJA2 Suppresses Colorectal Cancer Progression by Controlling HDAC2 Degradation and Stability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2401964. [PMID: 39928532 PMCID: PMC11967759 DOI: 10.1002/advs.202401964] [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: 02/24/2024] [Revised: 01/11/2025] [Indexed: 02/12/2025]
Abstract
PJA2 is documented to degrade various substrates. Nevertheless, the role of PJA2 as an E3 ubiquitin-protein ligase in colorectal cancer (CRC) progression remains unexplored. The correlation between PJA2 mRNA levels and clinical characteristics is investigated using data from The Cancer Genome Atlas (TCGA) database. Quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) are utilized to evaluate PJA2 expression levels in CRC tissues. The biological functions of PJA2 are confirmed through colony formation assays and azoxymethane/dextran sulfate sodium (AOM/DSS) mouse model of CRC, among other experimental approaches. The underlying molecular mechanisms of PJA2 action are elucidated using RNA sequencing (RNA-seq), co-immunoprecipitation (co-IP), proximity ligation assay (PLA), and chromatin immunoprecipitation (ChIP). Our research discovered that PJA2 is downregulated in CRC tissues and decreased PJA2 expression correlates with poor prognosis. Functionally, in vivo and in vitro experiments uncovered that PJA2 inhibits tumor cell proliferation and promotes apoptosis. Mechanistically, PJA2 recognized histone deacetylase 2 (HDAC2) via its RING-B-box domain (RBD) and bind to the N-terminal of HDAC2, facilitating ubiquitination at the lysine 90 (K90) residue. PJA2-mediated degradation of HDAC2 counteracts the transcriptional repression of the interferon-induced protein with the tetratricopeptide repeats (IFIT) family, thereby suppressing CRC progression. The data demonstrates that PJA2 suppresses CRC progression through the PJA2/HDAC2/IFIT axis, and its expression is regulated by HDAC2, thus constituting a positive feedback loop. Consequently, PJA2 may serve as a potential therapeutic target for CRC, and interrupting this feedback loop can represent a viable treatment strategy to restrain CRC progression.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Chaofan Peng
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Chi Jin
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Ye Wang
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Tuo Wang
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Peng Yang
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Wen Peng
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Qingyang Sun
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Hengjie Xu
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Hongxu Nie
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Xiaowei Wang
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Junwei Tang
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Yueming Sun
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| | - Yifei Feng
- Department of General SurgeryColorectal Institute of Nanjing Medical UniversityThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjingP. R. China
- The First School of Clinical MedicineNanjing Medical UniversityNanjingP. R. China
| |
Collapse
|
3
|
Liu B, Chen M, Liang Y, Mei Z, Sun W, Gao W, Zhang T, Wang R, Guo Y. ZC3H13 promotes autophagy in bladder cancer through m6A methylation modification of PJA2 and ubiquitination of KSR1. Hum Cell 2024; 38:23. [PMID: 39614918 DOI: 10.1007/s13577-024-01155-x] [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: 07/31/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025]
Abstract
The N6-methyladenine (m6A) modification is the most common modification of messenger RNAs in eukaryotes and has crucial roles in multiple cancers, including bladder cancer (BLCA). This paper aimed to probe the molecular mechanism of zinc-finger CCCH-type containing 13 (ZC3H13)-mediated N6-methyladenine (m6A) modification in BLCA progression via autophagy. Differential expression of ZC3H13 in BLCA was analyzed by the bioinformatics database. ZC3H13 expression in BLCA tissues and cell lines was determined, and malignant behaviors of BLCA cells were examined in vitro and in vivo. ZC3H13 was decreased in BLCA tissues and cell lines relative to adjacent tissues and normal uroepithelial cells. ZC3H13 overexpression restricted BLCA cell growth in vitro and curbed BLCA development in vivo. ZC3H13 promoted the mRNA stability of paraja ring finger 2 (PJA2) through m6A modification, leading to the ubiquitination degradation of the kinase suppressor of Ras 1 (KSR1). Knockdown of PJA2 and overexpression of KSR1 reversed the inhibitory effect of ZC3H13 on BLCA progression. ZC3H13 degraded KSR1 through m6A modification of PJA2, promoted cell autophagy, and repressed BLCA progression. Overall, ZC3H13 promotes the mRNA stability of PJA2 through m6A modification to degrade KSR1, thereby promoting autophagy in BLCA.
Collapse
Affiliation(s)
- Beibei Liu
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Mengjie Chen
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Yujie Liang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Zhijie Mei
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Wei Sun
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Wuyue Gao
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Tiantian Zhang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China
| | - Rui Wang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China.
| | - Yuanyuan Guo
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzi Lake District, Bengbu, 233004, Anhui, People's Republic of China.
| |
Collapse
|
4
|
Huang Y, Yang G, Yao X, Fang Y, Lin Q, Zhou M, Yang Y, Meng Q, Zhang Q, Wang S. Proteomic profiling of prostate cancer reveals molecular signatures under antiandrogen treatment. Clin Proteomics 2024; 21:44. [PMID: 38918720 PMCID: PMC11202386 DOI: 10.1186/s12014-024-09490-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Tumorigenesis and progression of prostate cancer (PCa) are indispensably dependent on androgen receptor (AR). Antiandrogen treatment is the principal preference for patients with advanced PCa. However, the molecular characteristics of PCa with antiandrogen intervention have not yet been fully uncovered. METHODS We first performed proteome analysis with 32 PCa tumor samples and 10 adjacent tissues using data-independent acquisition (DIA)- parallel accumulation serial fragmentation (PASEF) proteomics. Then label-free quantification (LFQ) mass spectrometry was employed to analyze protein profiles in LNCaP and PC3 cells. RESULTS M-type creatine kinase CKM and cartilage oligomeric matrix protein COMP were demonstrated to have the potential to be diagnostic biomarkers for PCa at both mRNA and protein levels. Several E3 ubiquitin ligases and deubiquitinating enzymes (DUBs) were significantly altered in PCa and PCa cells under enzalutamide treatment, and these proteins might reprogram proteostasis at protein levels in PCa. Finally, we discovered 127 significantly varied proteins in PCa samples with antiandrogen therapy and further uncovered 4 proteins in LNCaP cells upon enzalutamide treatment. CONCLUSIONS Our research reveals new potential diagnostic biomarkers for prostate cancer and might help resensitize resistance to antiandrogen therapy.
Collapse
Affiliation(s)
- Yurun Huang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Guanglin Yang
- Department of Urology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Xinpeng Yao
- The First Clinical Medical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Yue Fang
- The First Clinical Medical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Qiliang Lin
- The First Clinical Medical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Menghan Zhou
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Yiping Yang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Qinggui Meng
- Department of Urology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Qingyun Zhang
- Department of Urology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.
| | - Shan Wang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.
| |
Collapse
|
5
|
Schiefer S, Hale BG. Proximal protein landscapes of the type I interferon signaling cascade reveal negative regulation by PJA2. Nat Commun 2024; 15:4484. [PMID: 38802340 PMCID: PMC11130243 DOI: 10.1038/s41467-024-48800-5] [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: 10/03/2023] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Deciphering the intricate dynamic events governing type I interferon (IFN) signaling is critical to unravel key regulatory mechanisms in host antiviral defense. Here, we leverage TurboID-based proximity labeling coupled with affinity purification-mass spectrometry to comprehensively map the proximal human proteomes of all seven canonical type I IFN signaling cascade members under basal and IFN-stimulated conditions. This uncovers a network of 103 high-confidence proteins in close proximity to the core members IFNAR1, IFNAR2, JAK1, TYK2, STAT1, STAT2, and IRF9, and validates several known constitutive protein assemblies, while also revealing novel stimulus-dependent and -independent associations between key signaling molecules. Functional screening further identifies PJA2 as a negative regulator of IFN signaling via its E3 ubiquitin ligase activity. Mechanistically, PJA2 interacts with TYK2 and JAK1, promotes their non-degradative ubiquitination, and limits the activating phosphorylation of TYK2 thereby restraining downstream STAT signaling. Our high-resolution proximal protein landscapes provide global insights into the type I IFN signaling network, and serve as a valuable resource for future exploration of its functional complexities.
Collapse
Affiliation(s)
- Samira Schiefer
- Institute of Medical Virology, University of Zurich, 8057, Zurich, Switzerland
- Life Science Zurich Graduate School, ETH and University of Zurich, 8057, Zurich, Switzerland
| | - Benjamin G Hale
- Institute of Medical Virology, University of Zurich, 8057, Zurich, Switzerland.
| |
Collapse
|
6
|
Girik V, van Ek L, Dentand Quadri I, Azam M, Cruz Cobo M, Mandavit M, Riezman I, Riezman H, Gavin AC, Nunes-Hasler P. Development of Genetically Encoded Fluorescent KSR1-Based Probes to Track Ceramides during Phagocytosis. Int J Mol Sci 2024; 25:2996. [PMID: 38474242 DOI: 10.3390/ijms25052996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Ceramides regulate phagocytosis; however, their exact function remains poorly understood. Here, we sought (1) to develop genetically encoded fluorescent tools for imaging ceramides, and (2) to use them to examine ceramide dynamics during phagocytosis. Fourteen enhanced green fluorescent protein (EGFP) fusion constructs based on four known ceramide-binding domains were generated and screened. While most constructs localized to the nucleus or cytosol, three based on the CA3 ceramide-binding domain of kinase suppressor of ras 1 (KSR1) localized to the plasma membrane or autolysosomes. C-terminally tagged CA3 with a vector-based (C-KSR) or glycine-serine linker (C-KSR-GS) responded sensitively and similarly to ceramide depletion and accumulation using a panel of ceramide modifying drugs, whereas N-terminally tagged CA3 (N-KSR) responded differently to a subset of treatments. Lipidomic and liposome microarray analysis suggested that, instead, N-KSR may preferentially bind glucosyl-ceramide. Additionally, the three probes showed distinct dynamics during phagocytosis. Despite partial autolysosomal degradation, C-KSR and C-KSR-GS accumulated at the plasma membrane during phagocytosis, whereas N-KSR did not. Moreover, the weak recruitment of C-KSR-GS to the endoplasmic reticulum and phagosomes was enhanced through overexpression of the endoplasmic reticulum proteins stromal interaction molecule 1 (STIM1) and Sec22b, and was more salient in dendritic cells. The data suggest these novel probes can be used to analyze sphingolipid dynamics and function in living cells.
Collapse
Affiliation(s)
- Vladimir Girik
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Larissa van Ek
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Isabelle Dentand Quadri
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Maral Azam
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - María Cruz Cobo
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Marion Mandavit
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Isabelle Riezman
- Department of Biochemistry, NCCR Chemical Biology, Faculty of Science, University of Geneva, 1211 Geneva, Switzerland
| | - Howard Riezman
- Department of Biochemistry, NCCR Chemical Biology, Faculty of Science, University of Geneva, 1211 Geneva, Switzerland
| | - Anne-Claude Gavin
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Department of Biochemistry, NCCR Chemical Biology, Faculty of Science, University of Geneva, 1211 Geneva, Switzerland
| | - Paula Nunes-Hasler
- Department of Pathology and Immunology, Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
7
|
Rinaldi L, Chiuso F, Senatore E, Borzacchiello D, Lignitto L, Iannucci R, Donne RD, Fuggi M, Reale C, Russo F, Russo NA, Giurato G, Rizzo F, Sellitto A, Santangelo M, De Biase D, Paciello O, D'Ambrosio C, Amente S, Garbi C, Dalla E, Scaloni A, Weisz A, Ambrosino C, Insabato L, Feliciello A. Downregulation of praja2 restrains endocytosis and boosts tyrosine kinase receptors in kidney cancer. Commun Biol 2024; 7:208. [PMID: 38379085 PMCID: PMC10879500 DOI: 10.1038/s42003-024-05823-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 01/16/2024] [Indexed: 02/22/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common kidney cancer in the adult population. Late diagnosis, resistance to therapeutics and recurrence of metastatic lesions account for the highest mortality rate among kidney cancer patients. Identifying novel biomarkers for early cancer detection and elucidating the mechanisms underlying ccRCC will provide clues to treat this aggressive malignant tumor. Here, we report that the ubiquitin ligase praja2 forms a complex with-and ubiquitylates the AP2 adapter complex, contributing to receptor endocytosis and clearance. In human RCC tissues and cells, downregulation of praja2 by oncogenic miRNAs (oncomiRs) and the proteasome markedly impairs endocytosis and clearance of the epidermal growth factor receptor (EGFR), and amplifies downstream mitogenic and proliferative signaling. Restoring praja2 levels in RCC cells downregulates EGFR, rewires cancer cell metabolism and ultimately inhibits tumor cell growth and metastasis. Accordingly, genetic ablation of praja2 in mice upregulates RTKs (i.e. EGFR and VEGFR) and induces epithelial and vascular alterations in the kidney tissue.In summary, our findings identify a regulatory loop between oncomiRs and the ubiquitin proteasome system that finely controls RTKs endocytosis and clearance, positively impacting mitogenic signaling and kidney cancer growth.
Collapse
Affiliation(s)
- Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Domenica Borzacchiello
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Luca Lignitto
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille Univ, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Rosa Iannucci
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Rossella Delle Donne
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Mariano Fuggi
- Department of Advanced Biomedical Sciences, University Hospital Federico II, Naples, Italy
| | - Carla Reale
- Biogem, Biology and Molecular Genetics Institute, Ariano Irpino, Italy
| | - Filomena Russo
- Biogem, Biology and Molecular Genetics Institute, Ariano Irpino, Italy
| | | | - Giorgio Giurato
- Genome Research Center for Health, Baronissi (SA), Italy
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, Baronissi (SA), Italy
| | - Francesca Rizzo
- Genome Research Center for Health, Baronissi (SA), Italy
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, Baronissi (SA), Italy
| | - Assunta Sellitto
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, Baronissi (SA), Italy
| | - Michele Santangelo
- Department of Advanced Biomedical Sciences, University Hospital Federico II, Naples, Italy
| | - Davide De Biase
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, Pathology Unit, University Federico II, Naples, Italy
| | - Chiara D'Ambrosio
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici (Naples), Italy
| | - Stefano Amente
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Corrado Garbi
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Emiliano Dalla
- Department of Medicine, University of Udine, Udine, Italy
| | - Andrea Scaloni
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici (Naples), Italy
| | - Alessandro Weisz
- Genome Research Center for Health, Baronissi (SA), Italy
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, Baronissi (SA), Italy
| | - Concetta Ambrosino
- Biogem, Biology and Molecular Genetics Institute, Ariano Irpino, Italy
- Department of Science and Technology University of Sannio, Sannio, Italy
| | - Luigi Insabato
- Department of Advanced Biomedical Sciences, University Hospital Federico II, Naples, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy.
| |
Collapse
|
8
|
Onodera W, Kawasaki K, Oishi M, Aoki S, Asahi T. Functional Divergence and Origin of the Vertebrate Praja Family. J Mol Evol 2024; 92:21-29. [PMID: 38158403 DOI: 10.1007/s00239-023-10150-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
The Praja family is an E3 ubiquitin ligase, promoting polyubiquitination and subsequent degradation of substrates. It comprises two paralogs, praja1 and praja2. Prior research suggests these paralogs have undergone functional divergence, with examples, such as their distinct roles in neurite outgrowth. However, the specific evolutionary trajectories of each paralog remain largely unexplored preventing mechanistic understanding of functional differences between paralogs. Here, we investigated the phylogeny and divergence of the vertebrate Praja family through molecular evolutionary analysis. Phylogenetic examination of the vertebrate praja revealed that praja1 and praja2 originated from the common ancestor of placentals via gene duplication, with praja1 evolving at twice the rate of praja2 shortly after the duplication. Moreover, a unique evolutionary trajectory for praja1 relative to other vertebrate Praja was indicated, as evidenced by principal component analysis on GC content, codon usage frequency, and amino acid composition. Subsequent motif/domain comparison revealed conserved N terminus and C terminus in praja1 and praja2, together with praja1-specific motifs, including nuclear localization signal and Ala-Gly-Ser repeats. The nuclear localization signal was demonstrated to be functional in human neuroblastoma SH-SY5Y cells using deletion mutant, while praja2 was exclusively expressed in the nucleus. These discoveries contribute to a more comprehensive understanding of the Praja family's phylogeny and suggest a functional divergence between praja1 and praja2. Specifically, the shift of praja1 into the nucleus implies the degradation of novel substrates located in the nucleus as an evolutionary consequence.
Collapse
Affiliation(s)
- Wataru Onodera
- Faculty of Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu, Shinjuku, Tokyo, 162-0056, Japan.
| | - Kotaro Kawasaki
- Faculty of Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu, Shinjuku, Tokyo, 162-0056, Japan
| | - Mizuho Oishi
- Faculty of Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu, Shinjuku, Tokyo, 162-0056, Japan
| | - Shiho Aoki
- Faculty of Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu, Shinjuku, Tokyo, 162-0056, Japan
| | - Toru Asahi
- Faculty of Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu, Shinjuku, Tokyo, 162-0056, Japan.
- Research Organization for Nano & Life Innovation, Waseda University, 513 Waseda-Tsurumaki, Shinjuku, Tokyo, 162-0041, Japan.
| |
Collapse
|
9
|
Chiuso F, Delle Donne R, Giamundo G, Rinaldi L, Borzacchiello D, Moraca F, Intartaglia D, Iannucci R, Senatore E, Lignitto L, Garbi C, Conflitti P, Catalanotti B, Conte I, Feliciello A. Ubiquitylation of BBSome is required for ciliary assembly and signaling. EMBO Rep 2023; 24:e55571. [PMID: 36744302 PMCID: PMC10074118 DOI: 10.15252/embr.202255571] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/27/2022] [Accepted: 01/17/2023] [Indexed: 02/07/2023] Open
Abstract
Bardet-Biedl syndrome (BBS) is a ciliopathy characterized by retinal degeneration, obesity, renal abnormalities, postaxial polydactyly, and developmental defects. Genes mutated in BBS encode for components and regulators of the BBSome, an octameric complex that controls the trafficking of cargos and receptors within the primary cilium. Although both structure and function of the BBSome have been extensively studied, the impact of ubiquitin signaling on BBSome is largely unknown. We identify the E3 ubiquitin ligase PJA2 as a novel resident of the ciliary compartment and regulator of the BBSome. Upon GPCR-cAMP stimulation, PJA2 ubiquitylates BBSome subunits. We demonstrate that ubiquitylation of BBS1 at lysine 143 increases the stability of the BBSome and promotes its binding to BBS3, an Arf-like GTPase protein controlling the targeting of the BBSome to the ciliary membrane. Downregulation of PJA2 or expression of a ubiquitylation-defective BBS1 mutant (BBS1K143R ) affects the trafficking of G-protein-coupled receptors (GPCRs) and Shh-dependent gene transcription. Expression of BBS1K143R in vivo impairs cilium formation, embryonic development, and photoreceptors' morphogenesis, thus recapitulating the BBS phenotype in the medaka fish model.
Collapse
Affiliation(s)
- Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Rossella Delle Donne
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Giuliana Giamundo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Biology, University of Naples Federico II, Naples, Italy
| | - Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Domenica Borzacchiello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Federica Moraca
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy.,Net4Science srl, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | | | - Rosa Iannucci
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Luca Lignitto
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy.,Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille Univ, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Corrado Garbi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Paolo Conflitti
- Faculty of Biomedical Sciences, Institute of Computational Science, Università della Svizzera Italiana (USI), Lugano, Switzerland
| | - Bruno Catalanotti
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Biology, University of Naples Federico II, Naples, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
10
|
Delle Donne R, Iannucci R, Rinaldi L, Roberto L, Oliva MA, Senatore E, Borzacchiello D, Lignitto L, Giurato G, Rizzo F, Sellitto A, Chiuso F, Castaldo S, Scala G, Campani V, Nele V, De Rosa G, D'Ambrosio C, Garbi C, Scaloni A, Weisz A, Ambrosino C, Arcella A, Feliciello A. Targeted inhibition of ubiquitin signaling reverses metabolic reprogramming and suppresses glioblastoma growth. Commun Biol 2022; 5:780. [PMID: 35918402 PMCID: PMC9345969 DOI: 10.1038/s42003-022-03639-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 06/27/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most frequent and aggressive form of primary brain tumor in the adult population; its high recurrence rate and resistance to current therapeutics urgently demand a better therapy. Regulation of protein stability by the ubiquitin proteasome system (UPS) represents an important control mechanism of cell growth. UPS deregulation is mechanistically linked to the development and progression of a variety of human cancers, including GBM. Thus, the UPS represents a potentially valuable target for GBM treatment. Using an integrated approach that includes proteomics, transcriptomics and metabolic profiling, we identify praja2, a RING E3 ubiquitin ligase, as the key component of a signaling network that regulates GBM cell growth and metabolism. Praja2 is preferentially expressed in primary GBM lesions expressing the wild-type isocitrate dehydrogenase 1 gene (IDH1). Mechanistically, we found that praja2 ubiquitylates and degrades the kinase suppressor of Ras 2 (KSR2). As a consequence, praja2 restrains the activity of downstream AMP-dependent protein kinase in GBM cells and attenuates the oxidative metabolism. Delivery in the brain of siRNA targeting praja2 by transferrin-targeted self-assembling nanoparticles (SANPs) prevented KSR2 degradation and inhibited GBM growth, reducing the size of the tumor and prolonging the survival rate of treated mice. These data identify praja2 as an essential regulator of cancer cell metabolism, and as a potential therapeutic target to suppress GBM growth.
Collapse
Affiliation(s)
- Rossella Delle Donne
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Rosa Iannucci
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | | | | | - Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Domenica Borzacchiello
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Luca Lignitto
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, University of Salerno, Salerno, Italy
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, University of Salerno, Salerno, Italy
| | - Assunta Sellitto
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, University of Salerno, Salerno, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | | | - Giovanni Scala
- Department of Biology, University Federico II, Naples, Italy
| | | | - Valeria Nele
- Department of Pharmacy, University Federico II, Naples, Italy
| | | | - Chiara D'Ambrosio
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici (Naples), Italy
| | - Corrado Garbi
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Andrea Scaloni
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici (Naples), Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry SMS, University of Salerno, Salerno, Italy
- Genome Research Center for Health, Campus of Medicine, University of Salerno, Salerno, Italy
| | - Concetta Ambrosino
- Biogem, Ariano Irpino, Avellino, Italy
- Department of Science and Technology University of Sannio, Benevento, Italy
| | | | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy.
| |
Collapse
|
11
|
Kattan RE, Han H, Seo G, Yang B, Lin Y, Dotson M, Pham S, Menely Y, Wang W. Interactome analysis of human phospholipase D and phosphatidic acid-associated protein network. Mol Cell Proteomics 2022; 21:100195. [PMID: 35007762 PMCID: PMC8864472 DOI: 10.1016/j.mcpro.2022.100195] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/22/2021] [Accepted: 01/06/2022] [Indexed: 01/01/2023] Open
Abstract
Mammalian phospholipase D (PLD) enzyme family consists of six members. Among them, PLD1/2/6 catalyzes phosphatidic acid (PA) production, while PLD3/4/5 has no catalytic activities. Deregulation of the PLD-PA lipid signaling has been associated with various human diseases including cancer. However, a comprehensive analysis of the regulators and effectors for this crucial lipid metabolic pathway has not been fully achieved. Using a proteomic approach, we defined the protein interaction network for the human PLD family of enzymes and PA and revealed diverse cellular signaling events involving them. Through it, we identified PJA2 as a novel E3 ubiquitin ligase for PLD1 involved in control of the PLD1-mediated mammalian target of rapamycin signaling. Additionally, we showed that PA interacted with and positively regulated sphingosine kinase 1. Taken together, our study not only generates a rich interactome resource for further characterizing the human PLD-PA lipid signaling but also connects this important metabolic pathway with numerous biological processes. Defining the interactome of human phospholipase D enzymes and phosphatidic acid. PJA2 functions as an E3 ubiquitin ligase of phospholipase D1. Phosphatidic acid interacts with and positively regulates sphingosine kinase 1.
Collapse
Affiliation(s)
- Rebecca Elizabeth Kattan
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Han Han
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Gayoung Seo
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Bing Yang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Yongqi Lin
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Max Dotson
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Stephanie Pham
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Yahya Menely
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
12
|
Wang L, Liu X, Yue M, Liu Z, Zhang Y, Ma Y, Luo J, Li W, Bai J, Yao H, Chen Y, Li X, Feng D, Song X. Identification of hub genes in bladder cancer based on weighted gene co-expression network analysis from TCGA database. Cancer Rep (Hoboken) 2021; 5:e1557. [PMID: 34541834 PMCID: PMC9458504 DOI: 10.1002/cnr2.1557] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/28/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022] Open
Abstract
Background Muscular invasive bladder cancer (MIBC) is a common malignant tumor in the world. Because of their heterogeneity in prognosis and response to treatment, biomarkers that can predict survival or help make treatment decisions in patients with MIBC are essential for individualized treatment. Aim We aimed to integrate bioinformatics research methods to identify a set of effective biomarkers capable of predicting, diagnosing, and treating MIBC. To provide a new theoretical basis for the diagnosis and treatment of bladder cancer. Methods and results Gene expression profiles and clinical data of MIBC were obtained by downloading from the Cancer Genome Atlas database. A dataset of 129 MIBC cases and controls was included. 2084 up‐regulated genes and 2961 down‐regulated genes were identified by differentially expressed gene (DEG) analysis. Then, gene ontology analysis was performed to explore the biological functions of DEGs, respectively. The up‐regulated DEGs are mainly enriched in epidermal cell differentiation, mitotic nuclear division, and so forth. They are also involved in the cell cycle, p53 signaling pathway, PPAR signaling pathway, and so forth. The weighted gene co‐expression network analysis yielded five modules related to pathological stages and grading, of which blue and turquoise were the most relevant modules for MIBC. Next, Using Kaplan–Meier survival analysis to identify further hub genes, the screening criteria at p ≤ .05, we found CNKSR1, HIP1R, CFL2, TPM1, CSRP1, SYNM, POPDC2, PJA2, and RBBP8NL genes associated with the progression and prognosis of MIBC patients. Finally, immunohistochemistry experiments further confirmed that CNKSR1 plays a vital role in the tumorigenic context of MIBC. Conclusion The research suggests that CNKSR1, POPDC2, and PJA2 may be novel biomarkers as therapeutic targets for MIBC, especially we used immunohistochemical further to validate CNKSR1 as a therapeutic target for MIBC which may help to improve the prognosis for MIBC.
Collapse
Affiliation(s)
- Lei Wang
- College of Life Sciences, Xinyang Normal University, Xinyang, China.,College of Life Medicine, Xinyang Normal University, Xinyang, China
| | - Xudong Liu
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Miao Yue
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Zhe Liu
- Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | - Yu Zhang
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Ying Ma
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Jia Luo
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Wuling Li
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Jiangshan Bai
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Hongmei Yao
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Yuxuan Chen
- Department of Recovery Medicine, People's Liberation Army 990 Hospital, Xinyang, China
| | - Xiaofeng Li
- Department of Pathology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinqiang Song
- College of Life Sciences, Xinyang Normal University, Xinyang, China.,College of Life Medicine, Xinyang Normal University, Xinyang, China
| |
Collapse
|
13
|
Zeng J, Zhang H, Tan Y, Wang Z, Li Y, Yang X. m6A demethylase FTO suppresses pancreatic cancer tumorigenesis by demethylating PJA2 and inhibiting Wnt signaling. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:277-292. [PMID: 34484859 PMCID: PMC8385122 DOI: 10.1016/j.omtn.2021.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 06/14/2021] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer is the deadliest malignancy of the digestive system and is the seventh most common cause of cancer-related deaths worldwide. The incidence and mortality of pancreatic cancer continue to increase, and its 5-year survival rate remains the lowest among all cancers. N6-methyladenine (m6A) is the most abundant reversible RNA modification in various eukaryotic messenger and long noncoding RNAs and plays crucial roles in the occurrence and development of cancers. However, the role of m6A in pancreatic cancer remains unclear. The present study aimed to explore the role of m6A and its regulators in pancreatic cancer and assess its underlying molecular mechanism associated with pancreatic cancer cell proliferation, invasion, and metastasis. Reduced expression of the m6A demethylase, fat mass and obesity-associated protein (FTO), was responsible for the high levels of m6A RNA modification in pancreatic cancer. Moreover, FTO demethylated the m6A modification of praja ring finger ubiquitin ligase 2 (PJA2), thereby reducing its mRNA decay, suppressing Wnt signaling, and ultimately restraining the proliferation, invasion, and metastasis of pancreatic cancer cells. Altogether, this study describes new, potential molecular therapeutic targets for pancreatic cancer that could pave the way to improve patient outcome.
Collapse
Affiliation(s)
- Juan Zeng
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang Liaoning 110004, China
| | - Heying Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang Liaoning 110004, China
| | - Yonggang Tan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang Liaoning 110004, China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang Liaoning 110004, China
| | - Yunwei Li
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang Liaoning 110004, China
| | - Xianghong Yang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang Liaoning 110004, China
| |
Collapse
|
14
|
Zheng L, Xu H, Di Y, Chen L, Liu J, Kang L, Gao L. ELK4 promotes the development of gastric cancer by inducing M2 polarization of macrophages through regulation of the KDM5A-PJA2-KSR1 axis. J Transl Med 2021; 19:342. [PMID: 34372882 PMCID: PMC8353876 DOI: 10.1186/s12967-021-02915-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 05/27/2021] [Indexed: 02/09/2023] Open
Abstract
Background We tried to elaborate the molecular mechanism of ETS-like transcription factor 4 (ELK4) affecting gastric cancer (GC) progression through M2 polarization of macrophages mediated by lysine-specific demethylase 5A (KDM5A)-Praja2 (PJA2)-kinase suppressor of ras 1 (KSR1) axis. Methods GC expression dataset was obtained from GEO database, and the downstream regulatory mechanism of ELK4 was predicted. Tumor-associated macrophages (TAMs) were isolated from GC tissues. The interaction among ELK4, KDM5A, PJA2 and KSR1 was analyzed by dual luciferase reporter gene, ChIP and Co-IP assays. The stability of KSR1 protein was detected by cycloheximide (CHX) treatment. After TAMs were co-cultured with HGC-27 cells, HGC-27 cell biological processes were assessed through gain- and loss-of function assays. Tumorigenicity was detected by tumorigenicity test in nude mice. Results In GC and TAMs, ELK4, KDM5A and KSR1 were highly expressed, while PJA2 was lowly expressed. M2 polarization of macrophages promoted the development of GC. ELK4 activated KDM5A by transcription and promoted macrophage M2 polarization. KDM5A inhibited the expression of PJA2 by removing H3K4me3 of PJA2 promoter, which promoted M2 polarization of macrophages. PJA2 reduced KSR1 by ubiquitination. ELK4 promoted the proliferative, migrative and invasive potentials of GC cells as well as the growth of GC xenografts by regulating KSR1. Conclusion ELK4 may reduce the PJA2-dependent inhibition of KSR1 by transcriptional activation of KDM5A to promote M2 polarization of macrophages, thus promoting the development of GC. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02915-1.
Collapse
Affiliation(s)
- Lei Zheng
- Department of Oncology, The First Hospital of Qinhuangdao, No. 258, Wenhua Road, Qinhuangdao, 066000, Hebei Province, People's Republic of China
| | - Hongmei Xu
- Department of Oncology, The First Hospital of Qinhuangdao, No. 258, Wenhua Road, Qinhuangdao, 066000, Hebei Province, People's Republic of China
| | - Ya Di
- Department of Oncology, The First Hospital of Qinhuangdao, No. 258, Wenhua Road, Qinhuangdao, 066000, Hebei Province, People's Republic of China
| | - Lanlan Chen
- Department of Oncology, The First Hospital of Qinhuangdao, No. 258, Wenhua Road, Qinhuangdao, 066000, Hebei Province, People's Republic of China
| | - Jiao Liu
- Department of Oncology, The First Hospital of Qinhuangdao, No. 258, Wenhua Road, Qinhuangdao, 066000, Hebei Province, People's Republic of China
| | - Liying Kang
- Department of Oncology, Tianjin Wuqing District People's Hospital, Tianjin, 301700, People's Republic of China
| | - Liming Gao
- Department of Oncology, The First Hospital of Qinhuangdao, No. 258, Wenhua Road, Qinhuangdao, 066000, Hebei Province, People's Republic of China.
| |
Collapse
|
15
|
Senatore E, Chiuso F, Rinaldi L, Intartaglia D, Delle Donne R, Pedone E, Catalanotti B, Pirone L, Fiorillo B, Moraca F, Giamundo G, Scala G, Raffeiner A, Torres-Quesada O, Stefan E, Kwiatkowski M, van Pijkeren A, Morleo M, Franco B, Garbi C, Conte I, Feliciello A. The TBC1D31/praja2 complex controls primary ciliogenesis through PKA-directed OFD1 ubiquitylation. EMBO J 2021; 40:e106503. [PMID: 33934390 PMCID: PMC8126939 DOI: 10.15252/embj.2020106503] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/03/2021] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
The primary cilium is a microtubule‐based sensory organelle that dynamically links signalling pathways to cell differentiation, growth, and development. Genetic defects of primary cilia are responsible for genetic disorders known as ciliopathies. Orofacial digital type I syndrome (OFDI) is an X‐linked congenital ciliopathy caused by mutations in the OFD1 gene and characterized by malformations of the face, oral cavity, digits and, in the majority of cases, polycystic kidney disease. OFD1 plays a key role in cilium biogenesis. However, the impact of signalling pathways and the role of the ubiquitin‐proteasome system (UPS) in the control of OFD1 stability remain unknown. Here, we identify a novel complex assembled at centrosomes by TBC1D31, including the E3 ubiquitin ligase praja2, protein kinase A (PKA), and OFD1. We show that TBC1D31 is essential for ciliogenesis. Mechanistically, upon G‐protein‐coupled receptor (GPCR)‐cAMP stimulation, PKA phosphorylates OFD1 at ser735, thus promoting OFD1 proteolysis through the praja2‐UPS circuitry. This pathway is essential for ciliogenesis. In addition, a non‐phosphorylatable OFD1 mutant dramatically affects cilium morphology and dynamics. Consistent with a role of the TBC1D31/praja2/OFD1 axis in ciliogenesis, alteration of this molecular network impairs ciliogenesis in vivo in Medaka fish, resulting in developmental defects. Our findings reveal a multifunctional transduction unit at the centrosome that links GPCR signalling to ubiquitylation and proteolysis of the ciliopathy protein OFD1, with important implications on cilium biology and development. Derangement of this control mechanism may underpin human genetic disorders.
Collapse
Affiliation(s)
- Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | | | - Rossella Delle Donne
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Emilia Pedone
- Institute of Biostructures and Bioimaging, CNR, Naples, Italy
| | | | - Luciano Pirone
- Institute of Biostructures and Bioimaging, CNR, Naples, Italy
| | - Bianca Fiorillo
- Department of Pharmacy, University Federico II, Naples, Italy
| | - Federica Moraca
- Department of Pharmacy, University Federico II, Naples, Italy.,Net4Science srl, University "Magna Graecia", Catanzaro, Italy
| | | | - Giovanni Scala
- Department of Biology, University Federico II, Naples, Italy
| | - Andrea Raffeiner
- Institute of Biochemistry, University of Innsbruck, Innsbruck, Austria.,Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Omar Torres-Quesada
- Institute of Biochemistry, University of Innsbruck, Innsbruck, Austria.,Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Eduard Stefan
- Institute of Biochemistry, University of Innsbruck, Innsbruck, Austria.,Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | | | | | - Manuela Morleo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Brunella Franco
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Science, University Federico II, Naples, Italy
| | - Corrado Garbi
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Biology, University Federico II, Naples, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| |
Collapse
|
16
|
Praja2 suppresses the growth of gastric cancer by ubiquitylation of KSR1 and inhibiting MEK-ERK signal pathways. Aging (Albany NY) 2021; 13:3886-3897. [PMID: 33461174 PMCID: PMC7906149 DOI: 10.18632/aging.202356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022]
Abstract
Gastric cancer (GC) is a common malignant tumor, which has a high incidence and fatality. Therefore, it is important to clarify the molecular mechanism of the occurrence and development for GC and to find more effective treatments and targeted drugs. In this study, we found that the kinase suppressor of Ras1 (KSR1) was increased in GC tissues and cell lines. Silencing of KSR1 inhibited the proliferation, migration and invasion of MKN-45 cells. E3 ligase Praja2 was downregulated in GC tissues and cell lines. In addition, praja2 promoted ubiquitylation of KSR1, but inhibited MEK-ERK signal pathways. Functional analysis indicated overexpression of praja2 inhibited the proliferation, migration and invasion of MKN-45 cells, while MG132 or FGF2 treatment removed the inhibitory effects of praja2 on GC progression. In vivo tumorigenesis experiments indicated praja2 inhibited tumor growth via KSR1-MEK-ERK axis. In conclusion, praja2 promoted the ubiquitylation and degradation of KSR1, which disturbed MEK- ERK signaling and inhibited GC progression. Our study might provide a novel target for GC clinical treatment.
Collapse
|
17
|
Scorziello A, Borzacchiello D, Sisalli MJ, Di Martino R, Morelli M, Feliciello A. Mitochondrial Homeostasis and Signaling in Parkinson's Disease. Front Aging Neurosci 2020; 12:100. [PMID: 32372945 PMCID: PMC7186467 DOI: 10.3389/fnagi.2020.00100] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
The loss of dopaminergic (DA) neurons in the substantia nigra leads to a progressive, long-term decline of movement and other non-motor deficits. The symptoms of Parkinson's disease (PD) often appear later in the course of the disease, when most of the functional dopaminergic neurons have been lost. The late onset of the disease, the severity of the illness, and its impact on the global health system demand earlier diagnosis and better targeted therapy. PD etiology and pathogenesis are largely unknown. There are mutations in genes that have been linked to PD and, from these complex phenotypes, mitochondrial dysfunction emerged as central in the pathogenesis and evolution of PD. In fact, several PD-associated genes negatively impact on mitochondria physiology, supporting the notion that dysregulation of mitochondrial signaling and homeostasis is pathogenically relevant. Derangement of mitochondrial homeostatic controls can lead to oxidative stress and neuronal cell death. Restoring deranged signaling cascades to and from mitochondria in PD neurons may then represent a viable opportunity to reset energy metabolism and delay the death of dopaminergic neurons. Here, we will highlight the relevance of dysfunctional mitochondrial homeostasis and signaling in PD, the molecular mechanisms involved, and potential therapeutic approaches to restore mitochondrial activities in damaged neurons.
Collapse
Affiliation(s)
- Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, University of Naples Federico II, Naples, Italy
| | - Domenica Borzacchiello
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Maria Jose Sisalli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, University of Naples Federico II, Naples, Italy
| | - Rossana Di Martino
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, University of Naples Federico II, Naples, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
18
|
Gong M, Ye S, Li WX, Zhang J, Liu Y, Zhu J, Lv W, Zhang H, Wang J, Lu A, He K. Regulatory function of praja ring finger ubiquitin ligase 2 mediated by the P2rx3/P2rx7 axis in mouse hippocampal neuronal cells. Am J Physiol Cell Physiol 2020; 318:C1123-C1135. [PMID: 32267716 DOI: 10.1152/ajpcell.00070.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Praja2 (Pja2), a member of the growing family of mammalian RING E3 ubiquitin ligases, is reportedly involved in not only several types of cancer but also neurological diseases and disorders, but the genetic mechanism underlying the regulation of Pja2 in the nervous system remains unclear. To study the cellular and molecular functions of Pja2 in mouse hippocampal neuronal cells (MHNCs), we used gain- and loss-of-function manipulations of Pja2 in HT-22 cells and tested their regulatory effects on three Alzheimer's disease (AD) genes and cell proliferation. The results revealed that the expression of AD markers, including amyloid beta precursor protein (App), microtubule-associated protein tau (Mapt), and gamma-secretase activating protein (Gsap), could be inhibited by Pja2 overexpression and activated by Pja2 knockdown. In addition, HT-22 cell proliferation was enhanced by Pja2 upregulation and suppressed by its downregulation. We also evaluated and quantified the targets that responded to the enforced expression of Pja2 by RNA-Seq, and the results showed that purinergic receptor P2X, ligand-gated ion channel 3 and 7 (P2rx3 and P2rx7), which show different expression patterns in the critical calcium signaling pathway, mediated the regulatory effect of Pja2 in HT-22 cells. Functional studies indicated that Pja2 regulated HT-22 cells development and AD marker genes by inhibiting P2rx3 but promoting P2rx7, a gene downstream of P2rx3. In conclusion, our results provide new insights into the regulatory function of the Pja2 gene in MHNCs and thus underscore the potential relevance of this molecule to the pathophysiology of AD.
Collapse
Affiliation(s)
- Mengting Gong
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Shoudong Ye
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, China.,Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Wen-Xing Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jian Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Yanjun Liu
- Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Jie Zhu
- Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Wenwen Lv
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Zhang
- Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Jing Wang
- Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Kan He
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, China.,Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, Anhui, China.,School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| |
Collapse
|
19
|
Combination Design of Time-Dependent Magnetic Field and Magnetic Nanocomposites to Guide Cell Behavior. NANOMATERIALS 2020; 10:nano10030577. [PMID: 32235724 PMCID: PMC7153399 DOI: 10.3390/nano10030577] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/11/2020] [Accepted: 03/17/2020] [Indexed: 12/11/2022]
Abstract
The concept of magnetic guidance is still challenging and has opened a wide range of perspectives in the field of tissue engineering. In this context, magnetic nanocomposites consisting of a poly(ε-caprolactone) (PCL) matrix and iron oxide (Fe3O4) nanoparticles were designed and manufactured for bone tissue engineering. The mechanical properties of PCL/Fe3O4 (80/20 w/w) nanocomposites were first assessed through small punch tests. The inclusion of Fe3O4 nanoparticles improved the punching properties as the values of peak load were higher than those obtained for the neat PCL without significantly affecting the work to failure. The effect of a time-dependent magnetic field on the adhesion, proliferation, and differentiation of human mesenchymal stem cells (hMSCs) was analyzed. The Alamar Blue assay, confocal laser scanning microscopy, and image analysis (i.e., shape factor) provided information on cell adhesion and viability over time, whereas the normalized alkaline phosphatase activity (ALP/DNA) demonstrated that the combination of a time-dependent field with magnetic nanocomposites (PCL/Fe3O4 Mag) influenced cell differentiation. Furthermore, in terms of extracellular signal-regulated kinase (ERK)1/2 phosphorylation, an insight into the role of the magnetic stimulation was reported, also demonstrating a strong effect due the combination of the magnetic field with PCL/Fe3O4 nanocomposites (PCL/Fe3O4 Mag).
Collapse
|
20
|
Mathur S, Fletcher AJ, Branigan E, Hay RT, Virdee S. Photocrosslinking Activity-Based Probes for Ubiquitin RING E3 Ligases. Cell Chem Biol 2019; 27:74-82.e6. [PMID: 31859248 PMCID: PMC6963778 DOI: 10.1016/j.chembiol.2019.11.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/13/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022]
Abstract
Activity-based protein profiling is an invaluable technique for studying enzyme biology and facilitating the development of therapeutics. Ubiquitin E3 ligases (E3s) are one of the largest enzyme families and regulate a host of (patho)physiological processes. The largest subtype are the RING E3s of which there are >600 members. RING E3s have adaptor-like activity that can be subject to diverse regulatory mechanisms and have become attractive drug targets. Activity-based probes (ABPs) for measuring RING E3 activity do not exist. Here we re-engineer ubiquitin-charged E2 conjugating enzymes to produce photocrosslinking ABPs. We demonstrate activity-dependent profiling of two divergent cancer-associated RING E3s, RNF4 and c-Cbl, in response to their native activation signals. We also demonstrate profiling of endogenous RING E3 ligase activation in response to epidermal growth factor (EGF) stimulation. These photocrosslinking ABPs should advance E3 ligase research and the development of selective modulators against this important class of enzymes. Photoactivated activity-based probes developed for large class of ubiquitin E3 ligases ABPs are compatible with divergent RING E3 activation mechanisms Parallelized E3 profiling and detection of growth factor-induced E3 activation
Collapse
Affiliation(s)
- Sunil Mathur
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK
| | - Adam J Fletcher
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK
| | - Emma Branigan
- Division of Gene Regulation and Expression, University of Dundee, Scotland, UK
| | - Ronald T Hay
- Division of Gene Regulation and Expression, University of Dundee, Scotland, UK
| | - Satpal Virdee
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Scotland, UK.
| |
Collapse
|
21
|
Frodyma D, Neilsen B, Costanzo-Garvey D, Fisher K, Lewis R. Coordinating ERK signaling via the molecular scaffold Kinase Suppressor of Ras. F1000Res 2017; 6:1621. [PMID: 29026529 PMCID: PMC5583734 DOI: 10.12688/f1000research.11895.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/01/2017] [Indexed: 12/17/2022] Open
Abstract
Many cancers, including those of the colon, lung, and pancreas, depend upon the signaling pathways induced by mutated and constitutively active Ras. The molecular scaffolds Kinase Suppressor of Ras 1 and 2 (KSR1 and KSR2) play potent roles in promoting Ras-mediated signaling through the Raf/MEK/ERK kinase cascade. Here we summarize the canonical role of KSR in cells, including its central role as a scaffold protein for the Raf/MEK/ERK kinase cascade, its regulation of various cellular pathways mediated through different binding partners, and the phenotypic consequences of KSR1 or KSR2 genetic inactivation. Mammalian KSR proteins have a demonstrated role in cellular and organismal energy balance with implications for cancer and obesity. Targeting KSR1 in cancer using small molecule inhibitors has potential for therapy with reduced toxicity to the patient. RNAi and small molecule screens using KSR1 as a reference standard have the potential to expose and target vulnerabilities in cancer. Interestingly, although KSR1 and KSR2 are similar in structure, KSR2 has a distinct physiological role in regulating energy balance. Although KSR proteins have been studied for two decades, additional analysis is required to elucidate both the regulation of these molecular scaffolds and their potent effect on the spatial and temporal control of ERK activation in health and disease.
Collapse
Affiliation(s)
- Danielle Frodyma
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Beth Neilsen
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Diane Costanzo-Garvey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Kurt Fisher
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Robert Lewis
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
22
|
Zhong J, Wang H, Chen W, Sun Z, Chen J, Xu Y, Weng M, Shi Q, Ma D, Miao C. Ubiquitylation of MFHAS1 by the ubiquitin ligase praja2 promotes M1 macrophage polarization by activating JNK and p38 pathways. Cell Death Dis 2017; 8:e2763. [PMID: 28471450 PMCID: PMC5520684 DOI: 10.1038/cddis.2017.102] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 01/18/2017] [Accepted: 02/13/2017] [Indexed: 02/07/2023]
Abstract
Sepsis is a systemic inflammation caused by infection. The balance between M1–M2 macrophage polarization has an essential role in the pathogenesis of sepsis. However, the exact mechanism underlying macrophage polarization is unclear. We previously showed that levels of malignant fibrous histiocytoma amplified sequence 1 (MFHAS1) were significantly elevated in septic patients compared with those in nonseptic patients, and involved in the activation of Toll-like receptor (TLR) 2/c-Jun N-terminal kinase (JNK)/nuclear factor (NF)-κB pathway. In the present study, we explored whether MFHAS1 was involved in macrophage polarization and determined the effect of MFHAS1 on inflammation. We performed in vitro pulldown assays and in vivo co-immunoprecipitation assays and found that E3 ubiquitin ligase praja2 could directly bind to MFHAS1. In situ immunostaining analysis confirmed the colocalization of endogenous praja2 with MFHAS1. We first reported that praja2 promotes the accumulation of ubiquitylated MFHAS1 but does not degrade it. Moreover, our results indicate that MFHAS1 ubiquitylation by praja2 positively regulates TLR2-mediated JNK/p38 pathway and promotes M1 macrophage polarization, M2 to M1 macrophage transformation and inflammation.
Collapse
Affiliation(s)
- Jing Zhong
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huihui Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, China
| | - Wankun Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhirong Sun
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiawei Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yajun Xu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Meilin Weng
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiqing Shi
- Department of Anesthesiology, Children's Hospital of Fudan University, Shanghai, China
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Collaborative Innovation Center of Genetics and Development, Institute of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Changhong Miao
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, China
| |
Collapse
|
23
|
PJA2 ubiquitinates the HIV-1 Tat protein with atypical chain linkages to activate viral transcription. Sci Rep 2017; 7:45394. [PMID: 28345603 PMCID: PMC5366948 DOI: 10.1038/srep45394] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 02/22/2017] [Indexed: 12/23/2022] Open
Abstract
Transcription complexes that assemble at the HIV-1 promoter efficiently initiate transcription but generate paused RNA polymerase II downstream from the start site. The virally encoded Tat protein hijacks positive transcription elongation factor b (P-TEFb) to phosphorylate and activate this paused polymerase. In addition, Tat undergoes a series of reversible post-translational modifications that regulate distinct steps of the transcription cycle. To identify additional functionally important Tat cofactors, we performed RNAi knockdowns of sixteen previously identified Tat interactors and found that a novel E3 ligase, PJA2, ubiquitinates Tat in a non-degradative manner and specifically regulates the step of HIV transcription elongation. Interestingly, several different lysine residues in Tat can function as ubiquitin acceptor sites, and variable combinations of these lysines support both full transcriptional activity and viral replication. Further, the polyubiquitin chain conjugated to Tat by PJA2 can itself be assembled through variable ubiquitin lysine linkages. Importantly, proper ubiquitin chain assembly by PJA2 requires that Tat first binds its P-TEFb cofactor. These results highlight that both the Tat substrate and ubiquitin modification have plastic site usage, and this plasticity is likely another way in which the virus exploits the host molecular machinery to expand its limited genetic repertoire.
Collapse
|