1
|
Peris-Díaz MD, Krężel A, Barran P. Deciphering the safeguarding role of cysteine residues in p53 against H 2O 2-induced oxidation using high-resolution native mass spectrometry. Commun Chem 2025; 8:13. [PMID: 39814824 PMCID: PMC11736120 DOI: 10.1038/s42004-024-01395-w] [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: 09/26/2024] [Accepted: 12/12/2024] [Indexed: 01/18/2025] Open
Abstract
The transcription factor p53 is exquisitely sensitive and selective to a broad variety of cellular environments. Several studies have reported that oxidative stress weakens the p53-DNA binding affinity for certain promoters depending on the oxidation mechanism. Despite this body of work, the precise mechanisms by which the physiologically relevant DNA-p53 tetramer complex senses cellular stresses caused by H2O2 are still unknown. Here, we employed native mass spectrometry (MS) and ion mobility (IM)-MS coupled to chemical labelling and H2O2-induced oxidation to examine the mechanism of redox regulation of the p53-p21 complex. Our approach has found that two reactive cysteines in p53 protect against H2O2-induced oxidation by forming reversible sulfenates.
Collapse
Affiliation(s)
- Manuel David Peris-Díaz
- Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, Manchester, UK.
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, F. Joliot-Curie 14a, Wrocław, Poland.
| | - Artur Krężel
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, F. Joliot-Curie 14a, Wrocław, Poland
| | - Perdita Barran
- Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, Manchester, UK.
| |
Collapse
|
2
|
Wang JR, Jurado-Aguilar J, Barroso E, Rodríguez-Calvo R, Camins A, Wahli W, Palomer X, Vázquez-Carrera M. PPARβ/δ upregulates the insulin receptor β subunit in skeletal muscle by reducing lysosomal activity and EphB4 levels. Cell Commun Signal 2024; 22:595. [PMID: 39696437 DOI: 10.1186/s12964-024-01972-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/30/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The increased degradation of the insulin receptor β subunit (InsRβ) in lysosomes contributes to the development of insulin resistance and type 2 diabetes mellitus. Endoplasmic reticulum (ER) stress contributes to insulin resistance through several mechanisms, including the reduction of InsRβ levels. Here, we examined how peroxisome proliferator-activated receptor (PPAR)β/δ regulates InsRβ levels in mouse skeletal muscle and C2C12 myotubes exposed to the ER stressor tunicamycin. METHODS Wild-type (WT) and Ppard-/- mice, WT mice treated with vehicle or the PPARβ/δ agonist GW501516, and C2C12 myotubes treated with the ER stressor tunicamycin or different activators or inhibitors were used. RESULTS Ppard-/- mice displayed reduced InsRβ protein levels in their skeletal muscle compared to wild-type (WT) mice, while the PPARβ/δ agonist GW501516 increased its levels in WT mice. Co-incubation of tunicamycin-exposed C2C12 myotubes with GW501516 partially reversed the decrease in InsRβ protein levels, attenuating both ER stress and the increase in lysosomal activity. In addition, the protein levels of the tyrosine kinase ephrin receptor B4 (EphB4), which binds to the InsRβ and facilitates its endocytosis and degradation in lysosomes, were increased in the skeletal muscle of Ppard-/- mice, with GW501516 reducing its levels in the skeletal muscle of WT mice. CONCLUSIONS Overall, these findings reveal that PPARβ/δ activation increases InsRβ levels by alleviating ER stress and lysosomal degradation.
Collapse
Affiliation(s)
- Jue-Rui Wang
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, 08028, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, 28029, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Javier Jurado-Aguilar
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, 08028, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, 28029, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, 08028, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, 28029, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Ricardo Rodríguez-Calvo
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, 28029, Spain
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, "Sant Joan" University Hospital, Universitat Rovira i Virgili, Pere Virgili Health Research Institute (IISPV), Reus, 43201, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, 08028, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, Lausanne, CH-1015, Switzerland
- INRAE ToxAlim, UMR1331, Chemin de Tournefeuille, Toulouse Cedex 3, F-31027, France
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, 08028, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, 28029, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, 08028, Spain.
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, 28029, Spain.
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain.
- Unitat de Farmacologia, Facultat de Farmàcia i Ciències de l'Alimentació, Av. Joan XXIII 27-31, Barcelona, E-08028, Spain.
| |
Collapse
|
3
|
Khanra S, Singh S, Singh TG. Mechanistic exploration of ubiquitination-mediated pathways in cerebral ischemic injury. Mol Biol Rep 2024; 52:22. [PMID: 39607439 DOI: 10.1007/s11033-024-10123-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
The ubiquitin-proteasome system (UPS) plays a pivotal role in regulating protein homeostasis and cellular processes, including protein degradation, trafficking, DNA repair, and cell signaling. During cerebral ischemia, ischemic conditions profoundly disrupt UPS activity, leading to proteasomal dysfunction and the accumulation of abnormal proteins. This imbalance contributes to neuronal injury and cell death observed in ischemic stroke. The UPS is intricately linked to various signaling pathways crucial for neuronal survival, inflammation, and cellular stress response, such as NF-κB, TRIM, TRIP, JAK-STAT, PI3K/Akt, and ERK1/2. Alterations in the ubiquitination process can significantly impact the activation and regulation of these pathways, exacerbating ischemic brain injury. Therapeutic approaches targeting the UPS in cerebral ischemia aim to rebalance protein levels, reduce proteotoxic stress, and mitigate neuronal injury. Strategies include proteasome inhibition, targeting specific ubiquitin ligases and deubiquitinating enzymes, and modulating ubiquitination-mediated regulation of key signaling pathways implicated in ischemia-induced pathophysiology. Therefore, the present review discusses the molecular mechanisms underlying UPS dysfunction in ischemic stroke is crucial for developing effective therapeutic interventions. Modulating ubiquitination-mediated pathways through therapeutic interventions targeting specific UPS components holds significant promise for mitigating ischemic brain injury and promoting neuroprotection and functional recovery in patients with cerebral ischemia.
Collapse
Affiliation(s)
- Supriya Khanra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| |
Collapse
|
4
|
Sadiq A, Fert-Bober J. PAD inhibition downregulates the cellular fibrotic behavior of senescent myofibroblasts derived from dilated cardiomyopathy. Biomed Pharmacother 2024; 180:117579. [PMID: 39442233 DOI: 10.1016/j.biopha.2024.117579] [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: 08/13/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is characterized by enlarged, weakened heart ventricles due to chronic fibrosis. Dysfunctional senescent myofibroblasts and excessive citrullination have been implicated in fibrotic diseases. Peptidylarginine deiminases (PADs) are involved in the citrullination of ECM proteins. However, their role in regulating the cellular functions of cardiac myofibroblasts in DCM, is not well understood. This study aimed to evaluate the role of PADs in the cellular biology and fibrotic behavior of myofibroblasts in DCM. RESULTS Aged cardiac myofibroblasts derived from dilated cardiomyopathy (DCM, N=5) and healthy (HCF, N=3) participants (35-60 years), were cultured in TGFB-conditioned medium and treated with an irreversible pan-PAD inhibitor BB-Cl-amidine. Our findings showed that, compared with HCFs, DCM myofibroblasts showed high expression of PAD-2, PAD-3, citrullinated proteins and ECM proteins (vimentin, fibronectin, actin, and b-Tubulin). BB-Cl-amidine-mediated PAD inhibition directly affected the cell biology of DCM myofibroblasts, as shown by the reduced migration and invasion of DCM myofibroblasts. It also augmented the apoptosis by activating caspase-3 and decreased senescence by regulating p-53. PAD inhibition did not affect the citrullination of vimentin or fibronectin; however, it decreased collagen 1 A expression. CONCLUSIONS This study revealed that elevated PAD expression facilitates cellular processes mainly senescence, migration, and invasion. PAD inhibition resulted in the downregulation of these cellular functions, thereby reducing the fibrotic behavior of DCM myofibroblasts.
Collapse
Affiliation(s)
- Alia Sadiq
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Advanced Clinical Biosystems Research Institute, Precision Biomarker Laboratories, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Justyna Fert-Bober
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Advanced Clinical Biosystems Research Institute, Precision Biomarker Laboratories, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Tang J, Lam GT, Brooks RD, Miles M, Useckaite Z, Johnson IR, Ung BSY, Martini C, Karageorgos L, Hickey SM, Selemidis S, Hopkins AM, Rowland A, Vather R, O'Leary JJ, Brooks DA, Caruso MC, Logan JM. Exploring the role of sporadic BRAF and KRAS mutations during colorectal cancer pathogenesis: A spotlight on the contribution of the endosome-lysosome system. Cancer Lett 2024; 585:216639. [PMID: 38290660 DOI: 10.1016/j.canlet.2024.216639] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 02/01/2024]
Abstract
The highly heterogenous nature of colorectal cancer can significantly hinder its early and accurate diagnosis, eventually contributing to high mortality rates. The adenoma-carcinoma sequence and serrated polyp-carcinoma sequence are the two most common sequences in sporadic colorectal cancer. Genetic alterations in adenomatous polyposis coli (APC), v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) and tumour protein 53 (TP53) genes are critical in adenoma-carcinoma sequence, whereas v-Raf murine sarcoma viral oncogene homolog B (BRAF) and MutL Homolog1 (MLH1) are driving oncogenes in the serrated polyp-carcinoma sequence. Sporadic mutations in these genes contribute differently to colorectal cancer pathogenesis by introducing distinct alterations in several signalling pathways that rely on the endosome-lysosome system. Unsurprisingly, the endosome-lysosome system plays a pivotal role in the hallmarks of cancer and contributes to specialised colon function. Thus, the endosome-lysosome system might be distinctively influenced by different mutations and these alterations may contribute to the heterogenous nature of sporadic colorectal cancer. This review highlights potential connections between major sporadic colorectal cancer mutations and the diverse pathogenic mechanisms driven by the endosome-lysosome system in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Jingying Tang
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Giang T Lam
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Robert D Brooks
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Mark Miles
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Melbourne, Vic, Australia
| | - Zivile Useckaite
- College of Medicine and Public Health, Flinders University, Flinders Drive, Bedford Park, Adelaide, SA, Australia
| | - Ian Rd Johnson
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Ben S-Y Ung
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Carmela Martini
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Litsa Karageorgos
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Shane M Hickey
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Melbourne, Vic, Australia
| | - Ashley M Hopkins
- College of Medicine and Public Health, Flinders University, Flinders Drive, Bedford Park, Adelaide, SA, Australia
| | - Andrew Rowland
- College of Medicine and Public Health, Flinders University, Flinders Drive, Bedford Park, Adelaide, SA, Australia
| | - Ryash Vather
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, South Australia, Australia; Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Douglas A Brooks
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Maria C Caruso
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Jessica M Logan
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia.
| |
Collapse
|
6
|
Ren J, Yu P, Liu S, Li R, Niu X, Chen Y, Zhang Z, Zhou F, Zhang L. Deubiquitylating Enzymes in Cancer and Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303807. [PMID: 37888853 PMCID: PMC10754134 DOI: 10.1002/advs.202303807] [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: 06/11/2023] [Revised: 08/30/2023] [Indexed: 10/28/2023]
Abstract
Deubiquitylating enzymes (DUBs) maintain relative homeostasis of the cellular ubiquitome by removing the post-translational modification ubiquitin moiety from substrates. Numerous DUBs have been demonstrated specificity for cleaving a certain type of ubiquitin linkage or positions within ubiquitin chains. Moreover, several DUBs perform functions through specific protein-protein interactions in a catalytically independent manner, which further expands the versatility and complexity of DUBs' functions. Dysregulation of DUBs disrupts the dynamic equilibrium of ubiquitome and causes various diseases, especially cancer and immune disorders. This review summarizes the Janus-faced roles of DUBs in cancer including proteasomal degradation, DNA repair, apoptosis, and tumor metastasis, as well as in immunity involving innate immune receptor signaling and inflammatory and autoimmune disorders. The prospects and challenges for the clinical development of DUB inhibitors are further discussed. The review provides a comprehensive understanding of the multi-faced roles of DUBs in cancer and immunity.
Collapse
Affiliation(s)
- Jiang Ren
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
| | - Peng Yu
- Zhongshan Institute for Drug DiscoveryShanghai Institute of Materia MedicaChinese Academy of SciencesZhongshanGuangdongP. R. China
| | - Sijia Liu
- International Biomed‐X Research CenterSecond Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhouP. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhou310058China
| | - Ran Li
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
| | - Xin Niu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
| | - Yan Chen
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
| | - Zhenyu Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450003P. R. China
| | - Fangfang Zhou
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Long Zhang
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033P. R. China
- International Biomed‐X Research CenterSecond Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhouP. R. China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058P. R. China
| |
Collapse
|
7
|
Zhang J, Fang S, Rong F, Jia M, Wang Y, Cui H, Hao P. PSMD4 drives progression of hepatocellular carcinoma via Akt/COX2 pathway and p53 inhibition. Hum Cell 2023; 36:1755-1772. [PMID: 37336868 DOI: 10.1007/s13577-023-00935-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/12/2023] [Indexed: 06/21/2023]
Abstract
The ubiquitin-dependent proteolytic pathway is crucial for cellular regulation, including control of the cell cycle, differentiation, and apoptosis. Proteasome 26S Subunit Ubiquitin Receptor, Non-ATPase 4, (PSMD4) is a member of the ubiquitin proteasome family that is upregulated in multiple solid tumors, including hepatocellular carcinoma (HCC), and the existence of PSMD4 is associated with unfavorable prognosis. In this study, transcriptome sequencing of HCC tissues and non-tumor hepatic tissues from the public database Cancer Genome Atlas (TGCA) revealed a high expression of PSMD4. Additionally, PSMD4 loss in HCC cells suppressed the tumor development in mouse xenograft model. PSMD4, which is maintained by inflammatory factors secreted from tumor matrix cells, positively mediates cell growth and is associated with Akt/GSK-3β/ cyclooxygenase2 (COX2) pathway activation, inhibition of p53 promoter activity, and increased p53 degradation. However, the domain without the C-terminus (VWA+UIM1/2) sustained the activation of p53 transcription. Thus, our findings suggest that PSMD4 is involved in HCC tumor growth through COX2 expression and p53 downregulation. Therapeutic strategies targeting PSMD4 and its downstream effectors could be used for the treatment of PSMD4-abundant HCC patients.
Collapse
Affiliation(s)
- Jiamin Zhang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, Hebei, China
| | - Shu Fang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, Hebei, China
| | - Fanghao Rong
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, Hebei, China
| | - Miaomiao Jia
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, Hebei, China
| | - Yunpeng Wang
- Department of General Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Huixian Cui
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China.
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, Hebei, China.
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, Hebei, China.
| | - Peipei Hao
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China.
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, Hebei, China.
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, Hebei, China.
| |
Collapse
|
8
|
Müller A, Weyerhäuser P, Berte N, Jonin F, Lyubarskyy B, Sprang B, Kantelhardt SR, Salinas G, Opitz L, Schulz-Schaeffer W, Giese A, Kim EL. Concurrent Activation of Both Survival-Promoting and Death-Inducing Signaling by Chloroquine in Glioblastoma Stem Cells: Implications for Potential Risks and Benefits of Using Chloroquine as Radiosensitizer. Cells 2023; 12:cells12091290. [PMID: 37174691 PMCID: PMC10177603 DOI: 10.3390/cells12091290] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Lysosomotropic agent chloroquine was shown to sensitize non-stem glioblastoma cells to radiation in vitro with p53-dependent apoptosis implicated as one of the underlying mechanisms. The in vivo outcomes of chloroquine or its effects on glioblastoma stem cells have not been previously addressed. This study undertakes a combinatorial approach encompassing in vitro, in vivo and in silico investigations to address the relationship between chloroquine-mediated radiosensitization and p53 status in glioblastoma stem cells. Our findings reveal that chloroquine elicits antagonistic impacts on signaling pathways involved in the regulation of cell fate via both transcription-dependent and transcription-independent mechanisms. Evidence is provided that transcriptional impacts of chloroquine are primarily determined by p53 with chloroquine-mediated activation of pro-survival mevalonate and p21-DREAM pathways being the dominant response in the background of wild type p53. Non-transcriptional effects of chloroquine are conserved and converge on key cell fate regulators ATM, HIPK2 and AKT in glioblastoma stem cells irrespective of their p53 status. Our findings indicate that pro-survival responses elicited by chloroquine predominate in the context of wild type p53 and are diminished in cells with transcriptionally impaired p53. We conclude that p53 is an important determinant of the balance between pro-survival and pro-death impacts of chloroquine and propose that p53 functional status should be taken into consideration when evaluating the efficacy of glioblastoma radiosensitization by chloroquine.
Collapse
Affiliation(s)
- Andreas Müller
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Patrick Weyerhäuser
- Institute of Toxicology, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Nancy Berte
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Fitriasari Jonin
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Bogdan Lyubarskyy
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Bettina Sprang
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Sven Rainer Kantelhardt
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Gabriela Salinas
- NGS Integrative Genomics Core Unit (NIG), Institute for Human Genetics, University Medical Centre, 37075 Göttingen, Germany
| | - Lennart Opitz
- Functional Genomics Center Zurich, ETH Zurich, University of Zurich, 8092 Zurich, Switzerland
| | | | - Alf Giese
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Ella L Kim
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| |
Collapse
|
9
|
Liu X, Rossio V, Gygi SP, Paulo JA. Enriching Cysteine-Containing Peptides Using a Sulfhydryl-Reactive Alkylating Reagent with a Phosphonic Acid Group and Immobilized Metal Affinity Chromatography. J Proteome Res 2023; 22:1270-1279. [PMID: 36971515 PMCID: PMC10311885 DOI: 10.1021/acs.jproteome.2c00806] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
The reduction of disulfide bonds and their subsequent alkylation are commonplace in typical proteomics workflows. Here, we highlight a sulfhydryl-reactive alkylating reagent with a phosphonic acid group (iodoacetamido-LC-phosphonic acid, 6C-CysPAT) that facilitates the enrichment of cysteine-containing peptides for isobaric tag-based proteome abundance profiling. Specifically, we profile the proteome of the SH-SY5Y human cell line following 24 h treatments with two proteasome inhibitors (bortezomib and MG-132) in a tandem mass tag (TMT)pro9-plex experiment. We acquire three datasets─(1) Cys-peptide enriched, (2) the unbound complement, and (3) the non-depleted control─and compare the peptides and proteins quantified in each dataset, with emphasis on Cys-containing peptides. The data show that enrichment using 6C-Cys phosphonate adaptable tag (6C-CysPAT) can quantify over 38,000 Cys-containing peptides in 5 h with >90% specificity. In addition, our combined dataset provides the research community with a resource of over 9900 protein abundance profiles exhibiting the effects of two different proteasome inhibitors. Overall, the seamless incorporation of alkylation by 6C-CysPAT into a current TMT-based workflow permits the enrichment of a Cys-containing peptide subproteome. The acquisition of this "mini-Cys" dataset can be used to preview and assess the quality of a deep, fractionated dataset.
Collapse
Affiliation(s)
- Xinyue Liu
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Valentina Rossio
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
10
|
Chowdhury SR, Koley T, Singh M, Samath EA, Kaur P. Association of Hsp90 with p53 and Fizzy related homolog (Fzr) synchronizing Anaphase Promoting Complex (APC/C): An unexplored ally towards oncogenic pathway. Biochim Biophys Acta Rev Cancer 2023; 1878:188883. [PMID: 36972769 DOI: 10.1016/j.bbcan.2023.188883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/29/2023]
Abstract
The intricate molecular interactions leading to the oncogenic pathway are the consequence of cell cycle modification controlled by a bunch of cell cycle regulatory proteins. The tumor suppressor and cell cycle regulatory proteins work in coordination to maintain a healthy cellular environment. The integrity of this cellular protein pool is perpetuated by heat shock proteins/chaperones, which assist in proper protein folding during normal and cellular stress conditions. Among these versatile groups of chaperone proteins, Hsp90 is one of the significant ATP-dependent chaperones that aid in stabilizing many tumor suppressors and cell cycle regulator protein targets. Recently, studies have revealed that in cancerous cell lines, Hsp90 stabilizes mutant p53, 'the guardian of the genome.' Hsp90 also has a significant impact on Fzr, an essential regulator of the cell cycle having an important role in the developmental process of various organisms, including Drosophila, yeast, Caenorhabditis elegans, and plants. During cell cycle progression, p53 and Fzr coordinately regulate the Anaphase Promoting Complex (APC/C) from metaphase to anaphase transition up to cell cycle exit. APC/C mediates proper centrosome function in the dividing cell. The centrosome acts as the microtubule organizing center for the correct segregation of the sister chromatids to ensure perfect cell division. This review examines the structure of Hsp90 and its co-chaperones, which work in synergy to stabilize proteins such as p53 and Fizzy-related homolog (Fzr) to synchronize the Anaphase Promoting Complex (APC/C). Dysfunction of this process activates the oncogenic pathway leading to the development of cancer. Additionally, an overview of current drugs targeting Hsp90 at various phases of clinical trials has been included.
Collapse
Affiliation(s)
- Sanghati Roy Chowdhury
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Tirthankar Koley
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Mandeep Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
11
|
Li C, Lee H, Jung JH, Zhang Y, Wang J, Liu C, Sheffmaker RL, Segall AM, Zeng SX, Lu H. Coiled-coil domain containing 3 suppresses breast cancer growth by protecting p53 from proteasome-mediated degradation. Oncogene 2023; 42:154-164. [PMID: 36396725 DOI: 10.1038/s41388-022-02541-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022]
Abstract
Coiled-coil domain containing 3 (CCDC3) was previously shown to regulate liver lipid metabolism as a secretory protein. Here, we report an unexpected intracellular role of CCDC3 as a tumor suppressor in breast cancer (BrC). Bioinformatics datasets analysis showed that CCDC3 is under-expressed in BrCs, while its higher levels are correlated with higher overall survival and lower relapse of cancer patients, and CCDC3 is positively correlated with p53 and its target genes. Ectopic CCDC3 markedly suppressed proliferation, colony formation, and xenograft tumor growth by augmenting p53 activity in BrC cells. Depletion of endogenous CCDC3 by CRISPR-Cas9 increased proliferation and drug resistance of BrC cells by alleviating 5-Fluorouracil (5-FU)-induced p53 level and activity. Mechanistically, CCDC3 bound to the C-termini of p53 and MDM2, consequently stabilizing p53 in the nucleus and impairing MDM2 recruitment of p53 to the 26S proteosome without inhibiting p53 ubiquitination. p53 induced CCDC3 expression by binding to its promoter in BrC cells. Our results unveil a unique mechanism underlying CCDC3 activation of p53 in a positive feedback fashion to suppress BrC growth.
Collapse
Affiliation(s)
- Caiyue Li
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Hyemin Lee
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Ji Hoon Jung
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,College of Korean Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | - Yiwei Zhang
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Jieqiong Wang
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Chang Liu
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.,Department of Neuroscience, Tulane University, New Orleans, LA, 70118, USA
| | - Roger L Sheffmaker
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Allyson M Segall
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Shelya X Zeng
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA. .,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| | - Hua Lu
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA. .,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
12
|
Sparks A, Kelly CJ, Saville MK. Ubiquitin receptors play redundant roles in the proteasomal degradation of the p53 repressor MDM2. FEBS Lett 2022; 596:2746-2767. [PMID: 35735670 PMCID: PMC9796813 DOI: 10.1002/1873-3468.14436] [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: 05/11/2022] [Revised: 06/01/2022] [Accepted: 06/05/2022] [Indexed: 01/07/2023]
Abstract
Much remains to be determined about the participation of ubiquitin receptors in proteasomal degradation and their potential as therapeutic targets. Suppression of the ubiquitin receptor S5A/PSMD4/hRpn10 alone stabilises p53/TP53 but not the key p53 repressor MDM2. Here, we observed S5A and the ubiquitin receptors ADRM1/PSMD16/hRpn13 and RAD23A and B functionally overlap in MDM2 degradation. We provide further evidence that degradation of only a subset of ubiquitinated proteins is sensitive to S5A knockdown because ubiquitin receptor redundancy is commonplace. p53 can be upregulated by S5A modulation while degradation of substrates with redundant receptors is maintained. Our observations and analysis of Cancer Dependency Map (DepMap) screens show S5A depletion/loss substantially reduces cancer cell line viability. This and selective S5A dependency of proteasomal substrates make S5A a target of interest for cancer therapy.
Collapse
Affiliation(s)
| | - Christopher J. Kelly
- School of MedicineUniversity of DundeeUK,Institute of Infection, Immunity and InflammationUniversity of GlasgowUK
| | - Mark K. Saville
- School of MedicineUniversity of DundeeUK,Silver River EditingDundeeUK
| |
Collapse
|
13
|
Esfahanian N, Nelson M, Autenried R, Pattison JS, Callegari E, Rezvani K. Comprehensive Analysis of Proteasomal Complexes in Mouse Brain Regions Detects ENO2 as a Potential Partner of the Proteasome in the Striatum. Cell Mol Neurobiol 2022; 42:2305-2319. [PMID: 34037901 PMCID: PMC8617079 DOI: 10.1007/s10571-021-01106-2] [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: 02/11/2021] [Accepted: 05/19/2021] [Indexed: 11/28/2022]
Abstract
Defects in the activity of the proteasome or its regulators are linked to several pathologies, including neurodegenerative diseases. We hypothesize that proteasome heterogeneity and its selective partners vary across brain regions and have a significant impact on proteasomal catalytic activities. Using neuronal cell cultures and brain tissues obtained from mice, we compared proteasomal activities from two distinct brain regions affected in neurodegenerative diseases, the striatum and the hippocampus. The results indicated that proteasome activities and their responses to proteasome inhibitors are determined by their subcellular localizations and their brain regions. Using an iodixanol gradient fractionation method, proteasome complexes were isolated, followed by proteomic analysis for proteasomal interaction partners. Proteomic results revealed brain region-specific non-proteasomal partners, including gamma-enolase (ENO2). ENO2 showed more association to proteasome complexes purified from the striatum than to those from the hippocampus. These results highlight a potential key role for non-proteasomal partners of proteasomes regarding the diverse activities of the proteasome complex recorded in several brain regions.
Collapse
Affiliation(s)
- Niki Esfahanian
- Division of Basic Biomedical Sciences, Sanford School of Medicine,, University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA
| | - Morgan Nelson
- Division of Basic Biomedical Sciences, Sanford School of Medicine,, University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA
| | - Rebecca Autenried
- Division of Basic Biomedical Sciences, Sanford School of Medicine,, University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA
| | - J Scott Pattison
- Division of Basic Biomedical Sciences, Sanford School of Medicine,, University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA
| | - Eduardo Callegari
- Division of Basic Biomedical Sciences, Sanford School of Medicine,, University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA
| | - Khosrow Rezvani
- Division of Basic Biomedical Sciences, Sanford School of Medicine,, University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA.
| |
Collapse
|
14
|
Choi HS, Baek KH. Pro-apoptotic and anti-apoptotic regulation mediated by deubiquitinating enzymes. Cell Mol Life Sci 2022; 79:117. [PMID: 35118522 PMCID: PMC11071826 DOI: 10.1007/s00018-022-04132-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/20/2021] [Accepted: 01/05/2022] [Indexed: 12/16/2022]
Abstract
Although damaged cells can be repaired, cells that are considered unlikely to be repaired are eliminated through apoptosis, a type of predicted cell death found in multicellular organisms. Apoptosis is a structured cell death involving alterations to the cell morphology and internal biochemical changes. This process involves the expansion and cracking of cells, changes in cell membranes, nuclear fragmentation, chromatin condensation, and chromosome cleavage, culminating in the damaged cells being eaten and processed by other cells. The ubiquitin-proteasome system (UPS) is a major cellular pathway that regulates the protein levels through proteasomal degradation. This review proposes that apoptotic proteins are regulated through the UPS and describes a unique direction for cancer treatment by controlling proteasomal degradation of apoptotic proteins, and small molecules targeted to enzymes associated with UPS.
Collapse
Affiliation(s)
- Hae-Seul Choi
- Department of Biomedical Science, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi-Do, 13488, Republic of Korea.
| |
Collapse
|
15
|
Zhang S, Wang Y, Sun Y, Zhao G, Wang J, Liu L, Liu F, Wang P, Xu X. 4′,7-Di-O-methylnaringenin (DMNG), a naringenin derivative, activates p53 signal pathway through down-regulating MDM2. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.104962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
16
|
Huang T, Tian W, Zhou Q, Li J, Jiang Z, Chen J, Ge C, Tian H. Upregulation of Rpn10 promotes tumor progression via activation of the NF-κB pathway in clear cell renal cell carcinoma. Acta Biochim Biophys Sin (Shanghai) 2021; 53:988-996. [PMID: 34133712 DOI: 10.1093/abbs/gmab078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Indexed: 01/08/2023] Open
Abstract
The ubiquitin-proteasome system (UPS) plays a central role in regulating protein homeostasis in tumor progression. The proteasome subunit Rpn10 is associated with the progression of several tumor types. However, little is known regarding the role of Rpn10 in clear cell renal cell carcinoma (ccRCC). In this study, we found that overexpression of Rpn10 increased ccRCC cell proliferation, migration, and invasion. Silencing Rpn10 expression resulted in decreased cell proli-feration, migration, and invasion in ccRCC cells. Knockdown of Rpn10 inhibits tumor growth and cell proliferation in vivo. Furthermore, we demonstrated that Rpn10 increased cell proliferation, migration, and invasion via regulation of the nuclear factor kappa B (NF-κB) pathway. Rpn10 directly promoted inhibitor of nuclear factor-kappa B alpha (IκBα) degradation through the UPS. Moreover, we observed that upregulation of Rpn10 or downregulation of IκBα in ccRCC was associated with poor prognosis. We found that the combination of these two parameters was a more powerful predictor of poor prognosis than either parameter alone. Collectively, these findings provide evidence that Rpn10 promotes the progression of ccRCC by regulation of the NF-κB pathways and is a prognostic indicator for patients with ccRCC.
Collapse
Affiliation(s)
- Tingting Huang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Wei Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Qingqing Zhou
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Jiajun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Zhiyuan Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Jinsi Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Chao Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| |
Collapse
|
17
|
Wen J, Wang D. Deciphering the PTM codes of the tumor suppressor p53. J Mol Cell Biol 2021; 13:774-785. [PMID: 34289043 PMCID: PMC8782589 DOI: 10.1093/jmcb/mjab047] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 11/14/2022] Open
Abstract
The genome guardian p53 functions as a transcription factor that senses numerous cellular stresses and orchestrates the corresponding transcriptional events involved in determining various cellular outcomes, including cell cycle arrest, apoptosis, senescence, DNA repair, and metabolic regulation. In response to diverse stresses, p53 undergoes multiple posttranslational modifications (PTMs) that coordinate with intimate interdependencies to precisely modulate its diverse properties in given biological contexts. Notably, PTMs can recruit ‘reader’ proteins that exclusively recognize specific modifications and facilitate the functional readout of p53. Targeting PTM–reader interplay has been developing into a promising cancer therapeutic strategy. In this review, we summarize the advances in deciphering the ‘PTM codes’ of p53, focusing particularly on the mechanisms by which the specific reader proteins functionally decipher the information harbored within these PTMs of p53. We also highlight the potential applications of intervention with p53 PTM–reader interactions in cancer therapy and discuss perspectives on the ‘PTMomic’ study of p53 and other proteins.
Collapse
Affiliation(s)
- Jia Wen
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Donglai Wang
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
18
|
Zhang R, Cui D, Xue T, Lang Y, Zhang Y, Li L, Sun H, Kuang Y, Li G, Tang J. HLA-B-associated transcript 3 (Bat3) stabilizes and activates p53 in a HAUSP-dependent manner. J Mol Cell Biol 2021; 12:99-112. [PMID: 31647545 PMCID: PMC7109604 DOI: 10.1093/jmcb/mjz102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/12/2019] [Accepted: 08/26/2019] [Indexed: 01/06/2023] Open
Abstract
The p53 pathway is a highly complex signaling network including several key regulators. HAUSP is a critical component of the p53 pathway acting as a deubiquitinase for both p53 and its key repressor Mdm2. Here, we identified a novel HAUSP-interacting protein, HLA-B-associated transcript 3 (Bat3) and found it to be capable of inducing p53 stabilization and activation via a HAUSP-dependent mechanism, resulting in cell growth inhibition. Surprisingly, the deubiquitylating enzymatic activity of HAUSP was not required for this phenomenon. Co-immunoprecipitation showed that p53 coexisted in a complex with Bat3 and HAUSP in vivo, and HAUSP may serve as a binding mediator to enhance the interaction between p53 and Bat3. Further studies revealed that formation of this three-protein complex interfered with the binding of p53 to its proteasome receptor S5a and promoted the accumulation of p53 in nucleus. Notably, Mdm2 protein abundance is also regulated by Bat3 in the presence of HAUSP. Overexpression of Bat3 and HAUSP increases Mdm2 protein levels without influencing the p53–Mdm2 interaction and Mdm2-mediated p53 ubiquitination, indicating that Bat3–HAUSP-mediated protein stabilization is not specific to p53 and different mechanisms may be involved in Bat3-mediated regulation of p53–Mdm2 pathway. Together, our study unravels a novel mechanism by which p53 is stabilized and activated by HAUSP-mediated interaction with Bat3 and implies that Bat3 might function as a tumor suppressor through the stabilization of p53.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Basic Veterinary, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
| | - Di Cui
- Department of Basic Veterinary, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Teng Xue
- Department of Basic Veterinary, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
| | - Yue Lang
- Department of Basic Veterinary, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
| | - Yunfan Zhang
- Department of Basic Veterinary, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
| | - Lianjie Li
- Department of Basic Veterinary, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
| | - Haili Sun
- Department of Basic Veterinary, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
| | - Yu Kuang
- Department of Basic Veterinary, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Gebin Li
- Department of Clinical Veterinary, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jun Tang
- Department of Basic Veterinary, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
19
|
Good Cop, Bad Cop: Defining the Roles of Δ40p53 in Cancer and Aging. Cancers (Basel) 2020; 12:cancers12061659. [PMID: 32585821 PMCID: PMC7352174 DOI: 10.3390/cancers12061659] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 01/10/2023] Open
Abstract
The tumour suppressor p53 is essential for maintaining DNA integrity, and plays a major role in cellular senescence and aging. Understanding the mechanisms that contribute to p53 dysfunction can uncover novel possibilities for improving cancer therapies and diagnosis, as well as cognitive decline associated with aging. In recent years, the complexity of p53 signalling has become increasingly apparent owing to the discovery of the p53 isoforms. These isoforms play important roles in regulating cell growth and turnover in response to different stressors, depending on the cellular context. In this review, we focus on Δ40p53, an N-terminally truncated p53 isoform. Δ40p53 can alter p53 target gene expression in both a positive and negative manner, modulating the biological outcome of p53 activation; it also functions independently of p53. Therefore, proper control of the Δ40p53: p53 ratio is essential for normal cell growth, aging, and responses to cancer therapy. Defining the contexts and the mechanisms by which Δ40p53 behaves as a "good cop or bad cop" is critical if we are to target this isoform therapeutically.
Collapse
|
20
|
Wu H, Sun H, He Z, Chen X, Li Y, Zhao X, Kong W, Kong W. The effect and mechanism of 19S proteasome PSMD11/Rpn6 subunit in D-Galactose induced mimetic aging models. Exp Cell Res 2020; 394:112093. [PMID: 32450067 DOI: 10.1016/j.yexcr.2020.112093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/14/2020] [Accepted: 05/16/2020] [Indexed: 11/30/2022]
Abstract
Regulating proteasome activity is a potent therapeutic aspect of age-related hearing loss, which has been proven to protect neurons from age-related damaging. PSMD11, subunit of the 19S proteasome regulatory particle, is known to mainly up-regulate proteasome activity and prolong aging. However, the mechanism of PSMD11 in age-related hearing loss has not been deeply explored. In the present study, we explore the function and mechanism of PSMD11 protecting neurons in d-Galactose (D-Gal) mimetic aging models. Age-related pathologies were detected by Taq-PCR, ABR, Transmission electron microscopy, toluidine blue and β-galactosidase staining. The relative expressions of the proteins were explored by Western blotting, oxyblot, immunoprecipitation and immunofluorescence. Flow cytometry was used to manifest the oxidative state. We discovered that proteasome activity was impaired with aging, and that ROS and toxic protein accumulated in D-Gal induced aging models. PSMD11 changed with aging, and was associated with the metabolism of proteasome activity in the D-Gal treated models. Moreover, the knockdown or overexpression of PSMD11 was sufficient to change the oxidative state caused by D-Gal. Our results also demonstrated that PSMD11 could bond to AMPKα1/2 in the auditory cortex and PC12 cells, and AMPKα2 but not AMPKα1 was efficient to regulate the function of PSMD11. Deeper insights into the mechanisms of regulating PSMD11 for the anti-aging process are needed, and may offer novel therapeutic methods for central presbycusis.
Collapse
Affiliation(s)
- Han Wu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Haiying Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zuhong He
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xi Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yongqin Li
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xueyan Zhao
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Wen Kong
- Departments of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| | - Weijia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
21
|
Zhang L, Xu H, Ma C, Zhang J, Zhao Y, Yang X, Wang S, Li D. Upregulation of deubiquitinase PSMD14 in lung adenocarcinoma (LUAD) and its prognostic significance. J Cancer 2020; 11:2962-2971. [PMID: 32226511 PMCID: PMC7086243 DOI: 10.7150/jca.39539] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 01/24/2020] [Indexed: 01/01/2023] Open
Abstract
PSMD14 is a 19S-proteasome-associated deubiquitinating enzyme that facilitates protein degradation by the 20S proteasome core particle. Although accumulating evidence indicates that PSMD14 has emerged as a critical oncogenic factor by promoting tumor growth, the expression and function of PSMD14 in non-small cell lung cancer (NSCLC) remain largely unknown. In this study, we assessed PSMD14 expression and correlated it with clinical-pathological features and patient survival in NSCLC. We also determined the roles of PSMD14 in the regulation of lung adenocarcinoma (LUAD) cell growth. The results showed that PSMD14 expression was significantly upregulated in human NSCLC tissues compared with adjacent non-cancerous tissues. The PSMD14 level was associated with tumor size, lymph node invasion, and TNM stage in LUAD patients. Importantly, high PSMD14 expression was associated with poor overall survival (OS) and disease-free survival (DFS) in LUAD patients. Further, knockdown of PSMD14 significantly inhibited cell growth and caused G1 arrest and cellular senescence by increasing p21 stability in LUAD cells. PSMD14 knockdown also promoted cell apoptosis by increasing cleaved caspase-3 levels in H1299 cells. PSMD14 may serve as a potential prognostic marker and therapeutic target for LUAD patients.
Collapse
Affiliation(s)
- Ling Zhang
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, 68 Jiyang W Rd, Suzhou, 215600, China
| | - Hui Xu
- Department of Thoracic Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, 68 Jiyang W Rd, Suzhou, 215600, China
| | - Chunping Ma
- Department of Thoracic Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, 68 Jiyang W Rd, Suzhou, 215600, China
| | - Jieru Zhang
- Department of Respiratory & Critical Care Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, 68 Jiyang W Rd, Suzhou, 215600, China
| | - Yuanjie Zhao
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, 68 Jiyang W Rd, Suzhou, 215600, China
| | - Xiaomei Yang
- Department of Emergency, The Affiliated Zhangjiagang Hospital of Soochow University, 68 Jiyang W Rd, Suzhou, 215600, China
| | - Shusheng Wang
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, 68 Jiyang W Rd, Suzhou, 215600, China
| | - Dawei Li
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, 68 Jiyang W Rd, Suzhou, 215600, China
| |
Collapse
|
22
|
Cordani M, Butera G, Pacchiana R, Masetto F, Mullappilly N, Riganti C, Donadelli M. Mutant p53-Associated Molecular Mechanisms of ROS Regulation in Cancer Cells. Biomolecules 2020; 10:biom10030361. [PMID: 32111081 PMCID: PMC7175157 DOI: 10.3390/biom10030361] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 12/16/2022] Open
Abstract
The TP53 tumor suppressor gene is the most frequently altered gene in tumors and an increasing number of studies highlight that mutant p53 proteins can acquire oncogenic properties, referred to as gain-of-function (GOF). Reactive oxygen species (ROS) play critical roles as intracellular messengers, regulating numerous signaling pathways linked to metabolism and cell growth. Tumor cells frequently display higher ROS levels compared to healthy cells as a result of their increased metabolism as well as serving as an oncogenic agent because of its damaging and mutational properties. Several studies reported that in contrast with the wild type protein, mutant p53 isoforms fail to exert antioxidant activities and rather increase intracellular ROS, driving a pro-tumorigenic survival. These pro-oxidant oncogenic abilities of GOF mutant p53 include signaling and metabolic rewiring, as well as the modulation of critical ROS-related transcription factors and antioxidant systems, which lead ROS unbalance linked to tumor progression. The studies summarized here highlight that GOF mutant p53 isoforms might constitute major targets for selective therapeutic intervention against several types of tumors and that ROS enhancement driven by mutant p53 might represent an “Achilles heel” of cancer cells, suggesting pro-oxidant drugs as a therapeutic approach for cancer patients bearing the mutant TP53 gene.
Collapse
Affiliation(s)
- Marco Cordani
- IMDEA Nanociencia, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain;
| | - Giovanna Butera
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (R.P.); (F.M.); (N.M.)
| | - Raffaella Pacchiana
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (R.P.); (F.M.); (N.M.)
| | - Francesca Masetto
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (R.P.); (F.M.); (N.M.)
| | - Nidula Mullappilly
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (R.P.); (F.M.); (N.M.)
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10126 Torino, Italy;
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (R.P.); (F.M.); (N.M.)
- Correspondence: ; Tel.: +39-045-8027281; Fax: +39-045-8027170
| |
Collapse
|
23
|
Urolithin A induces prostate cancer cell death in p53-dependent and in p53-independent manner. Eur J Nutr 2019; 59:1607-1618. [DOI: 10.1007/s00394-019-02016-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/28/2019] [Indexed: 11/27/2022]
|
24
|
Inhibition of PSMD4 blocks the tumorigenesis of hepatocellular carcinoma. Gene 2019; 702:66-74. [DOI: 10.1016/j.gene.2019.03.063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/14/2019] [Accepted: 03/27/2019] [Indexed: 02/07/2023]
|
25
|
Lee B, Min JA, Nashed A, Lee SO, Yoo JC, Chi SW, Yi GS. A novel mechanism of irinotecan targeting MDM2 and Bcl-xL. Biochem Biophys Res Commun 2019; 514:518-523. [PMID: 31056264 DOI: 10.1016/j.bbrc.2019.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 11/26/2022]
Abstract
Irinotecan is a strong anticancer drug whose mechanism of action has been reported only for the inhibition of DNA topoisomerase I (Topo I) through its active metabolite SN-38. In this study, we present a new mechanism of Irinotecan which inhibits the activities of MDM2, an E3 ligase of tumour suppressor p53, and Bcl-xL, an anti-apoptotic protein, through direct binding. In our structure modelling study, Irinotecan could fit to the binding sites of MDM2 and Bcl-xL for their known drugs, Nutlin-3 and ABT-737, with a better binding affinity than to Topo I. The direct binding of Irinotecan to both proteins was confirmed through a NMR study. We further showed that Irinotecan increased the amount of p53 only in the presence of MDM2 and inhibited the physical interaction of Bcl-xL with Bim, a core pro-apoptotic protein. In addition, we demonstrated that Irinotecan induced the down regulation of proliferation and strong G2/M arrest in HCT116 colon cancer cells shortly after treatment. Collectively, we suggest a new mechanism of action for Irinotecan as a dual target inhibitor of MDM2 and Bcl-xL facilitating the anticancer activities mediated by p53 and Bcl-xL interaction partners.
Collapse
Affiliation(s)
- Boah Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea
| | - Jeong A Min
- Disease Target Structure Research Center, KRIBB, Daejeon, 34141, South Korea; Department of Proteome Structural Biology, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, South Korea
| | - Abdullateef Nashed
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea
| | - Sang-Ok Lee
- Disease Target Structure Research Center, KRIBB, Daejeon, 34141, South Korea
| | - Jae Cheal Yoo
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea
| | - Seung-Wook Chi
- Disease Target Structure Research Center, KRIBB, Daejeon, 34141, South Korea; Department of Proteome Structural Biology, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, South Korea.
| | - Gwan-Su Yi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, South Korea.
| |
Collapse
|
26
|
Expression of MDM2 in Macrophages Promotes the Early Postentry Steps of HIV-1 Infection through Inhibition of p53. J Virol 2019; 93:JVI.01871-18. [PMID: 30674627 DOI: 10.1128/jvi.01871-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/13/2019] [Indexed: 12/19/2022] Open
Abstract
The molecular basis for HIV-1 susceptibility in primary human monocyte-derived macrophages (MDMs) was previously evaluated by comparing the transcriptome of infected and bystander populations. Careful analysis of the data suggested that the ubiquitin ligase MDM2 acted as a positive regulator of HIV-1 replication in MDMs. In this study, MDM2 silencing through transcript-specific small interfering RNAs in MDMs induced a reduction in HIV-1 reverse transcription and integration along with an increase in the expression of p53-induced genes, including CDKN1A Experiments with Nutlin-3, a pharmacological inhibitor of MDM2 p53-binding activity, showed a similar effect on HIV-1 infection, suggesting that the observed restriction in HIV-1 production results from the release/activation of p53 and not the absence of MDM2 per se Knockdown and inhibition of MDM2 also both correlate with a decrease in the Thr592-phosphorylated inactive form of SAMHD1. The expression level of MDM2 and the p53 activation status are therefore important factors in the overall susceptibility of macrophages to HIV-1 infection, bringing a new understanding of signaling events controlling the process of virus replication in this cell type.IMPORTANCE Macrophages, with their long life span in vivo and their resistance to HIV-1-mediated cytopathic effect, might serve as viral reservoirs, contributing to virus persistence in an infected individual. Identification of host factors that increase the overall susceptibility of macrophages to HIV-1 might provide new therapeutic targets for the efficient control of viral replication in these cells and limit the formation of reservoirs in exposed individuals. In this study, we demonstrate the importance of p53 regulation by MDM2, which creates a cellular environment more favorable to the early steps of HIV-1 replication. Moreover, we show that p53 stabilization reduces virus infection in human macrophages, highlighting the important role of p53 in antiviral immunity.
Collapse
|
27
|
Rpn10 promotes tumor progression by regulating hypoxia-inducible factor 1 alpha through the PTEN/Akt signaling pathway in hepatocellular carcinoma. Cancer Lett 2019; 447:1-11. [PMID: 30673593 DOI: 10.1016/j.canlet.2019.01.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/08/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022]
Abstract
The ubiquitin-proteasome pathway plays a pivotal role in tumor progression. Rpn10 is the major ubiquitin (Ub) receptor of the 26S proteasome. Mounting evidence shows that Rpn10 is associated with the progression of several tumor types. However, little is known regarding the mechanistic role of Rpn10 in hepatocellular carcinoma (HCC). In this study, we found that the upregulation of Rpn10 in HCC was associated with poor prognosis. The ectopic overexpression of Rpn10 increased HCC cell proliferation, whereas silencing Rpn10 expression resulted in decreased cell proliferation. Furthermore, we demonstrated that knockdown of Rpn10 induced cell cycle arrest at G1 phase in HCC cells. In addition, we found that Rpn10 increased cell proliferation via regulation of the PTEN/Akt pathways. Knockdown of Rpn10 induced suppression of cell proliferation could be reversed by overexpressing active Akt in HCC cells. Rpn10 directly promoted PTEN degradation through the ubiquitin-proteasome system. The transcription factor HIF1α directly bound to the Rpn10 promoter and increased its expression in HCC tissue. Moreover, we observed a significant correlation between HIF1α expression and Rpn10 levels in HCC patients and found that the combination of these two parameters was a more powerful predictor of poor prognosis than either parameter alone. Collectively, these findings highlight the molecular mechanism of Rpn10 expression in HCC and provide valuable information for cancer prognosis and treatment.
Collapse
|
28
|
Tan G, Qiu M, Chen L, Zhang S, Ke L, Liu J. JS-K, a nitric oxide pro-drug, regulates growth and apoptosis through the ubiquitin-proteasome pathway in prostate cancer cells. BMC Cancer 2017; 17:376. [PMID: 28549433 PMCID: PMC5446692 DOI: 10.1186/s12885-017-3351-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/12/2017] [Indexed: 01/17/2023] Open
Abstract
Background In view of the fact that JS-K might regulate ubiquitin E3 ligase and that ubiquitin E3 ligase plays an important role in the mechanism of CRPC formation, the goal was to investigate the probable mechanism by which JS-K regulates prostate cancer cells. Methods Proliferation inhibition by JS-K on prostate cancer cells was examined usingCCK-8 assays. Caspase 3/7 activity assays and flow cytometry were performed to examine whether JS-K induced apoptosis in prostate cancer cells. Western blotting and co-immunoprecipitation analyses investigated JS-K’s effects on the associated apoptosis mechanism. Real time-PCR and Western blotting were performed to assess JS-K’s effect on transcription of specific AR target genes. Western blotting was also performed to detect Siah2 and AR protein concentrations and co-immunoprecipitation to detect interactions of Siah2 and AR, NCoR1 and AR, and p300 and AR. Results JS-K inhibited proliferation and induced apoptosis in prostate cancer cells. JS-K increased p53 and Mdm2 concentrations and regulated the caspase cascade reaction-associated protein concentrations. JS-K inhibited transcription of AR target genes and down-regulated PSA protein concentrations. JS-K inhibited Siah2 interactions and also inhibited the ubiquitination of AR. With further investigation, JS-K was found to stabilize AR and NCoR1 interactions and diminish AR and p300 interactions. Conclusions The present results suggested that JS-K might have been able to inhibit proliferation and induce apoptosis via regulation of the ubiquitin-proteasome degradation pathway, which represented a promising platform for the development of new compounds for PCa treatments. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3351-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guobin Tan
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China
| | - Mingning Qiu
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China
| | - Lieqian Chen
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China
| | - Sai Zhang
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China
| | - Longzhi Ke
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China
| | - Jianjun Liu
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China.
| |
Collapse
|
29
|
Sulak M, Fong L, Mika K, Chigurupati S, Yon L, Mongan NP, Emes RD, Lynch VJ. TP53 copy number expansion is associated with the evolution of increased body size and an enhanced DNA damage response in elephants. eLife 2016; 5. [PMID: 27642012 PMCID: PMC5061548 DOI: 10.7554/elife.11994] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 09/17/2016] [Indexed: 12/21/2022] Open
Abstract
A major constraint on the evolution of large body sizes in animals is an increased risk of developing cancer. There is no correlation, however, between body size and cancer risk. This lack of correlation is often referred to as 'Peto's Paradox'. Here, we show that the elephant genome encodes 20 copies of the tumor suppressor gene TP53 and that the increase in TP53 copy number occurred coincident with the evolution of large body sizes, the evolution of extreme sensitivity to genotoxic stress, and a hyperactive TP53 signaling pathway in the elephant (Proboscidean) lineage. Furthermore, we show that several of the TP53 retrogenes (TP53RTGs) are transcribed and likely translated. While TP53RTGs do not appear to directly function as transcription factors, they do contribute to the enhanced sensitivity of elephant cells to DNA damage and the induction of apoptosis by regulating activity of the TP53 signaling pathway. These results suggest that an increase in the copy number of TP53 may have played a direct role in the evolution of very large body sizes and the resolution of Peto's paradox in Proboscideans. DOI:http://dx.doi.org/10.7554/eLife.11994.001 As time passes, healthy cells are more likely to become cancerous because more and more damaging mutations accumulate in the cell’s DNA. Assuming that all cells have a similar risk of acquiring mutations, larger and longer-lived animals – like elephants – should have a higher risk of cancer than smaller, shorter-lived animals – like mice. However, there does not appear to be any link between the size of an animal and its risk of developing cancer. Consequently, a key question in cancer biology is how very large animals protect themselves against these diseases. One gene that is often damaged during an animal’s lifetime is called TP53. This gene normally produces a tumor suppressor protein that senses when DNA is damaged or a cell is under stress and either briefly slows the cell’s growth while the damage is repaired or triggers cell death if the stress is overwhelming. One way that large animals could reduce their risk of cancer is to have extra copies of TP53 or other genes that encode tumor suppressor proteins. Here Sulak et al. used an evolutionary genomics approach to study TP53 in 61 animals of various sizes, including several large animals such as African elephants and Minke whales. All of the animals studied had at least one copy of TP53, and several had a few extra copies, known as TP53 retrogenes. African elephants – the largest living land mammal – had more retrogenes than any of the others with 19 in total. To investigate why African elephants have so many TP53 retrogenes, Sulak et al. also analyzed DNA from Asian elephants and several other closely related, but now extinct species, including the woolly mammoth. As expected, as species evolved larger body sizes they also evolved more TP53 retrogenes. Further experiments indicate that several of the TP53 retrogenes in African elephants are likely to be able to produce the tumor suppressor protein and that they contribute to elephant cells being better equipped to deal with DNA damage. The next step following on from this work will be to find out exactly how TP53 retrogenes help to protect animals from cancer. DOI:http://dx.doi.org/10.7554/eLife.11994.002
Collapse
Affiliation(s)
- Michael Sulak
- Department of Human Genetics, The University of Chicago, Chicago, United States
| | - Lindsey Fong
- Department of Human Genetics, The University of Chicago, Chicago, United States
| | - Katelyn Mika
- Department of Human Genetics, The University of Chicago, Chicago, United States
| | | | - Lisa Yon
- School of Veterinary Medicine and Science, University of Nottingham, Leicestershire, United Kingdom.,Faculty of Medicine and Health Sciences, University of Nottingham, Leicestershire, United Kingdom
| | - Nigel P Mongan
- School of Veterinary Medicine and Science, University of Nottingham, Leicestershire, United Kingdom.,Faculty of Medicine and Health Sciences, University of Nottingham, Leicestershire, United Kingdom.,Department of Pharmacology, Weill Cornell Medical College, New York, United States
| | - Richard D Emes
- School of Veterinary Medicine and Science, University of Nottingham, Leicestershire, United Kingdom.,Faculty of Medicine and Health Sciences, University of Nottingham, Leicestershire, United Kingdom.,Advanced Data Analysis Centre, University of Nottingham UK, Nottingham, United Kingdom
| | - Vincent J Lynch
- Department of Human Genetics, The University of Chicago, Chicago, United States
| |
Collapse
|
30
|
Wang DW, Peng ZJ, Ren GF, Wang GX. The different roles of selective autophagic protein degradation in mammalian cells. Oncotarget 2016; 6:37098-116. [PMID: 26415220 PMCID: PMC4741918 DOI: 10.18632/oncotarget.5776] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/31/2015] [Indexed: 01/01/2023] Open
Abstract
Autophagy is an intracellular pathway for bulk protein degradation and the removal of damaged organelles by lysosomes. Autophagy was previously thought to be unselective; however, studies have increasingly confirmed that autophagy-mediated protein degradation is highly regulated. Abnormal autophagic protein degradation has been associated with multiple human diseases such as cancer, neurological disability and cardiovascular disease; therefore, further elucidation of protein degradation by autophagy may be beneficial for protein-based clinical therapies. Macroautophagy and chaperone-mediated autophagy (CMA) can both participate in selective protein degradation in mammalian cells, but the process is quite different in each case. Here, we summarize the various types of macroautophagy and CMA involved in determining protein degradation. For this summary, we divide the autophagic protein degradation pathways into four categories: the post-translational modification dependent and independent CMA pathways and the ubiquitin dependent and independent macroautophagy pathways, and describe how some non-canonical pathways and modifications such as phosphorylation, acetylation and arginylation can influence protein degradation by the autophagy lysosome system (ALS). Finally, we comment on why autophagy can serve as either diagnostics or therapeutic targets in different human diseases.
Collapse
Affiliation(s)
- Da-wei Wang
- Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhen-ju Peng
- Medical Institute of Paediatrics, Qilu Children's Hospital of Shandong University, Jinan, Shandong, China
| | - Guang-fang Ren
- Medical Institute of Paediatrics, Qilu Children's Hospital of Shandong University, Jinan, Shandong, China
| | - Guang-xin Wang
- Medical Institute of Paediatrics, Qilu Children's Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
31
|
Lin PL, Chang JT, Wu DW, Huang CC, Lee H. Cytoplasmic localization of Nrf2 promotes colorectal cancer with more aggressive tumors via upregulation of PSMD4. Free Radic Biol Med 2016; 95:121-32. [PMID: 27033953 DOI: 10.1016/j.freeradbiomed.2016.03.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/08/2016] [Accepted: 03/16/2016] [Indexed: 12/16/2022]
Abstract
Differences in subcellular localization of Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) have been associated with poor outcomes in human cancers. However, the prognostic value of subcellular localization of Nrf2 in colorectal cancer and the underlying mechanism in tumor invasion remain unknown. We enrolled tumors from colorectal patients to evaluate Nrf2, NQO1, and HO-1 expression by immunohistochemistry. NQO1 and HO-1 positive tumors showed nearly complete expression of Nrf2 in the nucleus and/or showed partial expression in the nucleus/cytoplasm (nNrf2); however, tumors negative for NQO1 and HO-1 showed almost complete expression of Nrf2 in the cytoplasm and/or partial expression in the nucleus/cytoplasm (cNrf2). Kaplan-Meier and Cox regression analysis indicated poorer overall survival in patients with cNrf2 tumors than with nNrf2 tumors. Cell models provided evidence that cNrf2, rather than nNrf2, was responsible for cell invasion and soft agar growth triggered by activation of the NF-κB/AKT/β-catenin cascade. Mechanistically, cNrf2 persistently increased PSMD4 expression by the HIF1α/β-catenin axis, whereas PSMD4 reciprocally enhanced Nrf2 nuclear export by increasing CRM1 expression through p53 degradation. The mechanistic action of the cell model was further confirmed with a nude mouse animal model in which xenograft tumors induced by cNrf2 were nearly completely suppressed by the proteasomal inhibitor carfilzomib or the β-catenin inhibitor XAV939. We therefore suggest that PSMD4 or β-catenin might be potential targets for suppressing tumor aggressiveness, and consequently, improving outcomes in patients whose tumors express cNrf2.
Collapse
Affiliation(s)
- Po-Lin Lin
- Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC
| | - Jinghua Tsai Chang
- Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC
| | - De-Wei Wu
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan, ROC
| | - Chi-Chou Huang
- Department of Surgery, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan, ROC.
| |
Collapse
|
32
|
Xu Y, Jin J, Zhang W, Zhang Z, Gao J, Liu Q, Zhou C, Xu Q, Shi H, Hou Y, Shi J. EGFR/MDM2 signaling promotes NF-κB activation via PPARγ degradation. Carcinogenesis 2015; 37:215-222. [PMID: 26718225 DOI: 10.1093/carcin/bgv252] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 12/22/2015] [Indexed: 11/13/2022] Open
Abstract
Dysregulated expression of epidermal growth factor receptor (EGFR) has been implicated in many cancer events, while peroxisome proliferator-activated receptor γ (PPARγ) negatively regulates cancer progression. The molecular mechanism of EGFR interaction with PPARγ is still unclear. Here, we found that nuclear EGFR induced phosphorylation of PPARγ at Tyr-74 leading to PPARγ ubiquitination and degradation by mouse double minute 2 (MDM2) ubiquitin ligase. PPARγ degradation by EGFR/MDM2 signaling resulted in accumulation of nuclear factor-kappaB (NF-κB)/p65 protein levels and increasing NF-κB activation. In contrast, PPARγ-Y74A mutant reversed this event. Moreover, PPARγ-Y74A mutant suppressed cell proliferation and increased chemotherapeutic agent-induced cancer cell sensitivity. Importantly, the clinical findings show that the nuclear phosphorylation of PPARγ-Y74 and EGFR expression in colonic cancer tissues was higher than that in control normal tissues. Thus, our study revealed a novel molecular mechanism that nuclear EGFR/NF-κB signaling promoted cell proliferation by destructing PPARγ function, which provides a novel strategy for cancer treatment.
Collapse
Affiliation(s)
- Ying Xu
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University, Changzhou, Jiangsu 213037, People's Republic of China.,Institute of Life Science, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China.,Department of Central Laboratory and
| | - Jianhua Jin
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University , Changzhou, Jiangsu 213037 , People's Republic of China
| | - Wenbo Zhang
- Institute of Life Science, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China.,Department of General Surgery, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu 212002, People's Republic of China
| | - Zhi Zhang
- Institute of Life Science, Jiangsu University , Zhenjiang, Jiangsu 212013 , People's Republic of China
| | - Jiaming Gao
- Institute of Life Science, Jiangsu University , Zhenjiang, Jiangsu 212013 , People's Republic of China
| | - Qian Liu
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University , Changzhou, Jiangsu 213037 , People's Republic of China
| | - Chenglin Zhou
- Jiangsu Taizhou People's Hospital , Jiangsu 225309 , People's Republic of China
| | - Qinggang Xu
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University , Changzhou, Jiangsu 213037 , People's Republic of China
| | - Haifeng Shi
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University , Changzhou, Jiangsu 213037 , People's Republic of China
| | - Yongzhong Hou
- Department of Oncology, The Affiliated Wujin Hospital, Jiangsu University, Changzhou, Jiangsu 213037, People's Republic of China.,Institute of Life Science, Jiangsu University, Zhenjiang, Jiangsu 212013, People's Republic of China
| | - Juanjuan Shi
- Institute of Life Science, Jiangsu University , Zhenjiang, Jiangsu 212013 , People's Republic of China
| |
Collapse
|
33
|
An intrinsically disordered region of RPN10 plays a key role in restricting ubiquitin chain elongation in RPN10 monoubiquitination. Biochem J 2015. [DOI: 10.1042/bj20141571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The proteasomal ubiquitin receptor Rpn10 (regulatory particle non-ATPase 10) is monoubiquitinated by Rsp5 (reverses SPT-phenotype protein 5). We show that a disordered region flanking the ubiquitin-interacting motif of Rpn10 is required for restricting polyubiquitination in the process of Rpn10 monoubiquitination. A novel role of an unstructured protein domain in controlling ubiquitin chain elongation is proposed.
Collapse
|
34
|
Piterman R, Braunstein I, Isakov E, Ziv T, Navon A, Cohen S, Stanhill A. VWA domain of S5a restricts the ability to bind ubiquitin and Ubl to the 26S proteasome. Mol Biol Cell 2014; 25:3988-98. [PMID: 25318673 PMCID: PMC4263443 DOI: 10.1091/mbc.e13-11-0697] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The only stoichiometric proteasomal subunit found to reside outside the proteasome is the ubiquitin receptor S5a. S5a-dependent binding of substrates and shuttle factors is restricted to occur only on the proteasome, thus increasing efficiency of substrate degradation by the 26S proteasome. The 26S proteasome recognizes a vast number of ubiquitin-dependent degradation signals linked to various substrates. This recognition is mediated mainly by the stoichiometric proteasomal resident ubiquitin receptors S5a and Rpn13, which harbor ubiquitin-binding domains. Regulatory steps in substrate binding, processing, and subsequent downstream proteolytic events by these receptors are poorly understood. Here we demonstrate that mammalian S5a is present in proteasome-bound and free states. S5a is required for efficient proteasomal degradation of polyubiquitinated substrates and the recruitment of ubiquitin-like (Ubl) harboring proteins; however, S5a-mediated ubiquitin and Ubl binding occurs only on the proteasome itself. We identify the VWA domain of S5a as a domain that limits ubiquitin and Ubl binding to occur only upon proteasomal association. Multiubiquitination events within the VWA domain can further regulate S5a association. Our results provide a molecular explanation to how ubiquitin and Ubl binding to S5a is restricted to the 26S proteasome.
Collapse
Affiliation(s)
- Ravit Piterman
- Department of Biochemistry, Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Ilana Braunstein
- Department of Biochemistry, Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Elada Isakov
- Department of Biochemistry, Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Tamar Ziv
- Smoler Proteomics Center, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Ami Navon
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Shenhav Cohen
- Department of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Ariel Stanhill
- Department of Biochemistry, Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|