1
|
Mattoo S, Arora M, Sharma P, Pore SK. Targeting mammalian N-end rule pathway for cancer therapy. Biochem Pharmacol 2025; 231:116684. [PMID: 39613115 DOI: 10.1016/j.bcp.2024.116684] [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: 08/29/2024] [Revised: 11/12/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Regulated protein degradation plays a crucial role in maintaining proteostasis along with protein refolding and compartmentalisation which collectively control biological functions. The N-end rule pathway is a major ubiquitin-dependent protein degradation system. The short-lived protein substrates containing destabilizing amino acid residues (N-degrons) are recognized by E3 ubiquitin ligases containing UBR box domains (N-recognin) for degradation. The dysregulated pathway fails to maintain the metabolic stability of the substrate proteins which leads to diseases. The mammalian substrates of this pathway are involved in many hallmarks of cancer such as resisting cell death, evading growth suppression, chromosomal instability, angiogenesis, and deregulation of cellular metabolism. Besides, mutations in E3 N-recognin have been detected in human cancers. In this review, we discuss the mammalian N-end rule pathway components, functions, and mechanism of degradation of substrates, and their implications in cancer pathogenesis. We also discuss the impact of pharmacological and genetic inhibition of this pathway component on cancer cells and chemoresistance. We further highlight how this pathway can be manipulated for selective protein degradation; for instance, using PROTAC technique. The challenges and future perspectives to utilize this pathway as a drug target for cancer therapy are also discussed.
Collapse
Affiliation(s)
- Shria Mattoo
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida 201311, India
| | - Muskaan Arora
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida 201311, India
| | - Priyanka Sharma
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Noida 201311, India
| | - Subrata Kumar Pore
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida 201311, India.
| |
Collapse
|
2
|
Najafzadeh M, Sajjadi SM, Kharazi S, Karimifard F, Safarpour H, Kharazinejad E. Interactions between cancer and stroma mediated by extracellular vesicles. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:114. [DOI: 10.1186/s43042-024-00582-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/13/2024] [Indexed: 01/05/2025] Open
Abstract
AbstractExtracellular vehicles (EVs) are small membrane-bound particles that are released by both cancer and stromal cells. These vesicles have emerged as key mediators of intercellular communication within the tumor microenvironment. In particular, EVs have been shown to play a critical role in facilitating the interactions between cancer cells and the surrounding stroma. Through the transfer of various bioactive molecules, including proteins, lipids, and nucleic acids, EVs are able to modulate the behavior of recipient cells and promote tumorigenesis. Additionally, EVs can also contribute to the development of drug resistance and immune evasion, further highlighting their importance in cancer progression. This review will summarize the current knowledge regarding EV-mediated interactions between cancer and stromal cells, and discuss their implications for cancer diagnosis and therapy.
Collapse
|
3
|
Larsson P, Olsson M, Sarathchandra S, Fäldt Beding A, Forssell-Aronsson E, Kovács A, Karlsson P, Helou K, Parris TZ. Multi-omics analysis identifies repurposing bortezomib in the treatment of kidney-, nervous system-, and hematological cancers. Sci Rep 2024; 14:18576. [PMID: 39127727 PMCID: PMC11316778 DOI: 10.1038/s41598-024-62339-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 05/15/2024] [Indexed: 08/12/2024] Open
Abstract
Repurposing of FDA-approved drugs is a quick and cost-effective alternative to de novo drug development. Here, we identify genes involved in bortezomib sensitivity, predict cancer types that may benefit from treatment with bortezomib, and evaluate the mechanism-of-action of bortezomib in breast cancer (BT-474 and ZR-75-30), melanoma (A-375), and glioblastoma (A-172) cells in vitro. Cancer cell lines derived from cancers of the blood, kidney, nervous system, and skin were found to be significantly more sensitive to bortezomib than other organ systems. The in vitro studies confirmed that although bortezomib effectively inhibited the β5 catalytic site in all four cell lines, cell cycle arrest was only induced in G2/M phase and apoptosis in A-375 and A-172 after 24h. The genomic and transcriptomic analyses identified 33 genes (e.g. ALDH18A1, ATAD2) associated with bortezomib resistance. Taken together, we identified biomarkers predictive of bortezomib sensitivity and cancer types that might benefit from treatment with bortezomib.
Collapse
Affiliation(s)
- Peter Larsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Maxim Olsson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | | | - Anna Fäldt Beding
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Southern Älvsborg Hospital, Borås, Sweden
| | - Eva Forssell-Aronsson
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Khalil Helou
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Toshima Z Parris
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
4
|
Ferrito N, Báez-Flores J, Rodríguez-Martín M, Sastre-Rodríguez J, Coppola A, Isidoro-García M, Prieto-Matos P, Lacal J. Biomarker Landscape in RASopathies. Int J Mol Sci 2024; 25:8563. [PMID: 39201250 PMCID: PMC11354534 DOI: 10.3390/ijms25168563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 07/28/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
RASopathies are a group of related genetic disorders caused by mutations in genes within the RAS/MAPK signaling pathway. This pathway is crucial for cell division, growth, and differentiation, and its disruption can lead to a variety of developmental and health issues. RASopathies present diverse clinical features and pose significant diagnostic and therapeutic challenges. Studying the landscape of biomarkers in RASopathies has the potential to improve both clinical practices and the understanding of these disorders. This review provides an overview of recent discoveries in RASopathy molecular profiling, which extend beyond traditional gene mutation analysis. mRNAs, non-coding RNAs, protein expression patterns, and post-translational modifications characteristic of RASopathy patients within pivotal signaling pathways such as the RAS/MAPK, PI3K/AKT/mTOR, and Rho/ROCK/LIMK2/cofilin pathways are summarized. Additionally, the field of metabolomics holds potential for uncovering metabolic signatures associated with specific RASopathies, which are crucial for developing precision medicine. Beyond molecular markers, we also examine the role of histological characteristics and non-invasive physiological assessments in identifying potential biomarkers, as they provide evidence of the disease's effects on various systems. Here, we synthesize key findings and illuminate promising avenues for future research in RASopathy biomarker discovery, underscoring rigorous validation and clinical translation.
Collapse
Affiliation(s)
- Noemi Ferrito
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007 Salamanca, Spain; (N.F.); (J.B.-F.); (J.S.-R.); (A.C.)
- GIR of Biomedicine of Rare Diseases, University of Salamanca (USAL), 37007 Salamanca, Spain;
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
| | - Juan Báez-Flores
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007 Salamanca, Spain; (N.F.); (J.B.-F.); (J.S.-R.); (A.C.)
- GIR of Biomedicine of Rare Diseases, University of Salamanca (USAL), 37007 Salamanca, Spain;
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
| | - Mario Rodríguez-Martín
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007 Salamanca, Spain; (N.F.); (J.B.-F.); (J.S.-R.); (A.C.)
- GIR of Biomedicine of Rare Diseases, University of Salamanca (USAL), 37007 Salamanca, Spain;
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
| | - Julián Sastre-Rodríguez
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007 Salamanca, Spain; (N.F.); (J.B.-F.); (J.S.-R.); (A.C.)
| | - Alessio Coppola
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007 Salamanca, Spain; (N.F.); (J.B.-F.); (J.S.-R.); (A.C.)
- GIR of Biomedicine of Rare Diseases, University of Salamanca (USAL), 37007 Salamanca, Spain;
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
| | - María Isidoro-García
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
- Clinical Biochemistry Department, University Hospital of Salamanca, 37007 Salamanca, Spain
- Clinical Rare Diseases Reference Unit DiERCyL, 37007 Castilla y León, Spain
- Department of Medicine, University of Salamanca (USAL), 37007 Salamanca, Spain
| | - Pablo Prieto-Matos
- GIR of Biomedicine of Rare Diseases, University of Salamanca (USAL), 37007 Salamanca, Spain;
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Pediatrics, University Hospital of Salamanca, 37007 Salamanca, Spain
- Department of Biomedical and Diagnostics Science, University of Salamanca (USAL), 37007 Salamanca, Spain
| | - Jesus Lacal
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007 Salamanca, Spain; (N.F.); (J.B.-F.); (J.S.-R.); (A.C.)
- GIR of Biomedicine of Rare Diseases, University of Salamanca (USAL), 37007 Salamanca, Spain;
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
| |
Collapse
|
5
|
Gupta M, Johnson ANT, Cruz ER, Costa EJ, Guest RL, Li SHJ, Hart EM, Nguyen T, Stadlmeier M, Bratton BP, Silhavy TJ, Wingreen NS, Gitai Z, Wühr M. Global protein turnover quantification in Escherichia coli reveals cytoplasmic recycling under nitrogen limitation. Nat Commun 2024; 15:5890. [PMID: 39003262 PMCID: PMC11246515 DOI: 10.1038/s41467-024-49920-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 06/25/2024] [Indexed: 07/15/2024] Open
Abstract
Protein turnover is critical for proteostasis, but turnover quantification is challenging, and even in well-studied E. coli, proteome-wide measurements remain scarce. Here, we quantify the turnover rates of ~3200 E. coli proteins under 13 conditions by combining heavy isotope labeling with complement reporter ion quantification and find that cytoplasmic proteins are recycled when nitrogen is limited. We use knockout experiments to assign substrates to the known cytoplasmic ATP-dependent proteases. Surprisingly, none of these proteases are responsible for the observed cytoplasmic protein degradation in nitrogen limitation, suggesting that a major proteolysis pathway in E. coli remains to be discovered. Lastly, we show that protein degradation rates are generally independent of cell division rates. Thus, we present broadly applicable technology for protein turnover measurements and provide a rich resource for protein half-lives and protease substrates in E. coli, complementary to genomics data, that will allow researchers to study the control of proteostasis.
Collapse
Affiliation(s)
- Meera Gupta
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Alex N T Johnson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Edward R Cruz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Eli J Costa
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Randi L Guest
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | | | - Elizabeth M Hart
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Thao Nguyen
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Michael Stadlmeier
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Benjamin P Bratton
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Vanderbilt Institute of Infection, Immunology and Inflammation, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas J Silhavy
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Ned S Wingreen
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Zemer Gitai
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Martin Wühr
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
6
|
Negi H, Ravichandran A, Dasgupta P, Reddy S, Das R. Plasticity of the proteasome-targeting signal Fat10 enhances substrate degradation. eLife 2024; 13:e91122. [PMID: 38984715 PMCID: PMC11299979 DOI: 10.7554/elife.91122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 07/09/2024] [Indexed: 07/11/2024] Open
Abstract
The proteasome controls levels of most cellular proteins, and its activity is regulated under stress, quiescence, and inflammation. However, factors determining the proteasomal degradation rate remain poorly understood. Proteasome substrates are conjugated with small proteins (tags) like ubiquitin and Fat10 to target them to the proteasome. It is unclear if the structural plasticity of proteasome-targeting tags can influence substrate degradation. Fat10 is upregulated during inflammation, and its substrates undergo rapid proteasomal degradation. We report that the degradation rate of Fat10 substrates critically depends on the structural plasticity of Fat10. While the ubiquitin tag is recycled at the proteasome, Fat10 is degraded with the substrate. Our results suggest significantly lower thermodynamic stability and faster mechanical unfolding in Fat10 compared to ubiquitin. Long-range salt bridges are absent in the Fat10 structure, creating a plastic protein with partially unstructured regions suitable for proteasome engagement. Fat10 plasticity destabilizes substrates significantly and creates partially unstructured regions in the substrate to enhance degradation. NMR-relaxation-derived order parameters and temperature dependence of chemical shifts identify the Fat10-induced partially unstructured regions in the substrate, which correlated excellently to Fat10-substrate contacts, suggesting that the tag-substrate collision destabilizes the substrate. These results highlight a strong dependence of proteasomal degradation on the structural plasticity and thermodynamic properties of the proteasome-targeting tags.
Collapse
Affiliation(s)
- Hitendra Negi
- National Center for Biological Sciences, Tata Institute of Fundamental ResearchBangaloreIndia
- SASTRA University, ThirumalaisamudramThanjavurIndia
| | - Aravind Ravichandran
- National Center for Biological Sciences, Tata Institute of Fundamental ResearchBangaloreIndia
- SASTRA University, ThirumalaisamudramThanjavurIndia
| | - Pritha Dasgupta
- National Center for Biological Sciences, Tata Institute of Fundamental ResearchBangaloreIndia
| | - Shridivya Reddy
- National Center for Biological Sciences, Tata Institute of Fundamental ResearchBangaloreIndia
| | - Ranabir Das
- National Center for Biological Sciences, Tata Institute of Fundamental ResearchBangaloreIndia
| |
Collapse
|
7
|
Boccellato C, Rehm M. TRAIL-induced apoptosis and proteasomal activity - Mechanisms, signalling and interplay. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119688. [PMID: 38368955 DOI: 10.1016/j.bbamcr.2024.119688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/01/2024] [Accepted: 02/10/2024] [Indexed: 02/20/2024]
Abstract
Programmed cell death, in particular apoptosis, is essential during development and tissue homeostasis, and also is the primary strategy to induce cancer cell death by cytotoxic therapies. Precision therapeutics targeting TRAIL death receptors are being evaluated as novel anti-cancer agents, while in parallel highly specific proteasome inhibitors have gained approval as drugs. TRAIL-dependent signalling and proteasomal control of cellular proteostasis are intricate processes, and their interplay can be exploited to enhance therapeutic killing of cancer cells in combination therapies. This review provides detailed insights into the complex signalling of TRAIL-induced pathways and the activities of the proteasome. It explores their core mechanisms of action, pharmaceutical druggability, and describes how their interplay can be strategically leveraged to enhance cell death responses in cancer cells. Offering this comprehensive and timely overview will allow to navigate the complexity of the processes governing cell death mechanisms in TRAIL- and proteasome inhibitor-based treatment conditions.
Collapse
Affiliation(s)
- Chiara Boccellato
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart 70569, Germany.
| | - Markus Rehm
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart 70569, Germany; University of Stuttgart, Stuttgart Research Center Systems Biology, Stuttgart 70569, Germany.
| |
Collapse
|
8
|
Lobas AA, Saei AA, Lyu H, Zubarev RA, Gorshkov MV. Chemical Proteomics Reveals that the Anticancer Drug Everolimus Affects the Ubiquitin-Proteasome System. ACS Pharmacol Transl Sci 2024; 7:787-796. [PMID: 38481686 PMCID: PMC10928898 DOI: 10.1021/acsptsci.3c00316] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2025]
Abstract
Rapamycin is a natural antifungal, immunosuppressive, and antiproliferative compound that allosterically inhibits mTOR complex 1. The ubiquitin-proteasome system (UPS) responsible for protein turnover is usually not listed among the pathways affected by mTOR signaling. However, some previous studies have indicated the interplay between the UPS and mTOR. It has also been reported that rapamycin and its analogs can allosterically inhibit the proteasome itself. In this work, we studied the molecular effect of rapamycin and its analogs (rapalogs), everolimus and temsirolimus, on the A549 cell line by expression proteomics. The analysis of differentially expressed proteins showed that the cellular response to everolimus treatment is strikingly different from that to rapamycin and temsirolimus. In the cluster analysis, the effect of everolimus was similar to that of bortezomib, a well-established proteasome inhibitor. UPS-related pathways were enriched in the cluster of proteins specifically upregulated upon everolimus and bortezomib treatments, suggesting that both compounds have similar proteasome inhibition effects. In particular, the total amount of ubiquitin was significantly elevated in the samples treated with everolimus and bortezomib, and analysis of the polyubiquitination patterns revealed elevated intensities of the ubiquitin peptide with a GG modification at the K48 residue, consistent with a bottleneck in proteasomal protein degradation. Moreover, the everolimus treatment resulted in both ubiquitin phosphorylation and generation of a significant amount of semitryptic peptides, illustrating the increase in the protease activity. These observations suggest that everolimus affects the UPS in a unique way, and its mechanism of action is different from that of its close chemical analogs, rapamycin and temsirolimus.
Collapse
Affiliation(s)
- Anna A. Lobas
- V.
L. Talrose Institute for Energy Problems of Chemical Physics, Federal
Research Center for Chemical Physics, Russian
Academy of Sciences, 119334 Moscow, Russia
| | - Amir Ata Saei
- Division
of Physiological Chemistry I, Department of Medical Biochemistry and
Biophysics, Karolinska Institutet, SE-17 177 Stockholm, Sweden
- Biozentrum, University of Basel, 4056 Basel, Switzerland
- Center
for Translational Microbiome Research, Department of Microbiology,
Tumor and Cell Biology, Karolinska Institutet, SE-17 177 Stockholm, Sweden
| | - Hezheng Lyu
- Division
of Physiological Chemistry I, Department of Medical Biochemistry and
Biophysics, Karolinska Institutet, SE-17 177 Stockholm, Sweden
| | - Roman A. Zubarev
- Division
of Physiological Chemistry I, Department of Medical Biochemistry and
Biophysics, Karolinska Institutet, SE-17 177 Stockholm, Sweden
- The
National Medical Research Center for Endocrinology, 115478 Moscow, Russia
| | - Mikhail V. Gorshkov
- V.
L. Talrose Institute for Energy Problems of Chemical Physics, Federal
Research Center for Chemical Physics, Russian
Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
9
|
Park HC, Kim H, Kim JY, Lee HY, Lee J, Cha W, Ahn SH, Jeong WJ. PSMD1 as a prognostic marker and potential target in oropharyngeal cancer. BMC Cancer 2023; 23:1242. [PMID: 38104103 PMCID: PMC10725586 DOI: 10.1186/s12885-023-11689-2] [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: 07/30/2023] [Accepted: 11/28/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Despite the diverse genetic mutations in head and neck cancer, the chemotherapy outcome for this cancer has not improved for decades. It is urgent to select prognostic factors and therapeutic targets for oropharyngeal cancer to establish precision medicine. Recent studies have identified PSMD1 as a potential prognostic marker in several cancers. We aimed to assess the prognostic significance of PSMD1 expression in oropharyngeal squamous cell carcinoma (OPSCC) patients using immunohistochemistry. METHODS We studied 64 individuals with OPSCC tissue from surgery at Seoul National University Bundang Hospital between April 2008 and August 2017. Immunostaining analysis was conducted on the tissue microarray (TMA) sections (4 μm) for p16 and PSMD1. H-score, which scale from 0 to 300, was calculated from each nucleus, cytoplasm, and cellular expression. Clinicopathological data were compared with Chi-squared test, Fisher's exact test, t-test, and logistic regression. Survival data until 2021 were achieved from national statistical office of Korea. Kaplan-Meier method and cox-regression model were used for disease-specific survival (DSS) analysis. RESULTS H-score of 90 in nucleus was appropriate cutoff value for 'High PSMD1 expression' in OPSCC. Tonsil was more frequent location in low PSMD1 group (42/52, 80.8%) than in high PSMD1 group (4/12, 33.3%; P = .002). Early-stage tumor was more frequent in in low PSMD1 group (45/52, 86.5%) than in high PSMD1 group (6/12, 50%; P = .005). HPV was more positive in low PSMD1 group (43/52, 82.7%) than in high PSMD1 group (5/12, 41.7%; P = .016). Patients with PSMD1 high expression showed poorer DSS than in patients with PSMD1 low expression (P = .006 in log rank test). In multivariate analysis, PSMD1 expression, pathologic T staging, and specimen age were found to be associated with DSS (P = .011, P = .025, P = .029, respectively). CONCLUSIONS In our study, we established PSMD1 as a negative prognostic factor in oropharyngeal squamous cell carcinoma, indicating its potential as a target for targeted therapy and paving the way for future in vitro studies on drug repositioning.
Collapse
Affiliation(s)
- Hae Chan Park
- Department of Otorhinolaryngology-Head & Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hyojin Kim
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Ji-Yeong Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hye-Yeon Lee
- Department of Otorhinolaryngology-Head & Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jinyi Lee
- Department of Otorhinolaryngology-Head & Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - WonJae Cha
- Department of Otorhinolaryngology-Head & Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Soon-Hyun Ahn
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Woo-Jin Jeong
- Department of Otorhinolaryngology-Head & Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea.
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, Korea.
| |
Collapse
|
10
|
Meng Y, Hong C, Yang S, Qin Z, Yang L, Huang Y. Roles of USP9X in cellular functions and tumorigenesis (Review). Oncol Lett 2023; 26:506. [PMID: 37920433 PMCID: PMC10618932 DOI: 10.3892/ol.2023.14093] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/12/2023] [Indexed: 11/04/2023] Open
Abstract
Ubiquitin-specific peptidase 9X (USP9X) is involved in certain human diseases, including malignancies, atherosclerosis and certain diseases of the nervous system. USP9X promotes the deubiquitination and stabilization of diverse substrates, thereby exerting a versatile range of effects on pathological and physiological processes. USP9X serves vital roles in the processes of cell survival, invasion and migration in various types of cancer. The present review aims to highlight the current knowledge of USP9X in terms of its structure and the possible mediatory mechanisms involved in certain types of cancer, providing a thorough introduction to its biological functions in carcinogenesis and further outlining its oncogenic or suppressive properties in a diverse range of cancer types. Finally, several perspectives regarding USP9X-targeted pharmacological therapeutics in cancer development are discussed.
Collapse
Affiliation(s)
- Yimei Meng
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Chaojin Hong
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Sifu Yang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Zhiquan Qin
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Liu Yang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Yumei Huang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
11
|
Meneses-Urrea LA, Vaquero-Abellán M, Villegas Arenas D, Benachi Sandoval N, Hernández-Carrillo M, Molina-Recio G. Association between Cervical Cancer and Dietary Patterns in Colombia. Nutrients 2023; 15:4889. [PMID: 38068749 PMCID: PMC10707912 DOI: 10.3390/nu15234889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Cervical cancer is a global public health problem. It is the second leading cause of death among women of childbearing age worldwide. Several factors, including diet, have been shown to influence the risk of persistent HPV infection and tumor progression. This paper determines the relationship between dietary patterns and cervical cancer. It is an ecological study of multiple groups, based on two national sources: the High-Cost Account and the National Survey of Nutritional Situation of Colombia of 2015. The population consisted of 3472 women aged 35 to 64. The incidence of cervical cancer was used as the dependent variable while the independent variables included food consumption according to established patterns, area of residence, age, physical activity, and BMI, among other variables. The statistical analysis performed through associations between variables was evaluated by multiple linear regression using R2. 38.9% of the evaluated population belonged to the first quartile of wealth, and 76.5% resided in the municipal capital. The incidence of cervical cancer in Colombia was associated with being affiliated to a state-subsidized health regime and having diabetes mellitus. A conservative eating pattern, as well as belonging to a rural area, are evidenced as protective factors. These results invite the need to encourage public policies and promote healthy lifestyles.
Collapse
Affiliation(s)
- Luz Adriana Meneses-Urrea
- Research Group “Health Care (Recognized by Colciencias)”, Universidad Santiago de Cali, Cali 760001, Colombia; (L.A.M.-U.); (D.V.A.); (N.B.S.)
- Department of Nursing, Universidad Santiago de Cali, Cali 760001, Colombia
| | - Manuel Vaquero-Abellán
- IMIBIC GC12 Clinical and Epidemiological Research in Primary Care (GICEAP), 14014 Córdoba, Spain
- Department of Nursing, Pharmacology and Physiotherapy, University of Córdoba, 14014 Córdoba, Spain;
| | - Dolly Villegas Arenas
- Research Group “Health Care (Recognized by Colciencias)”, Universidad Santiago de Cali, Cali 760001, Colombia; (L.A.M.-U.); (D.V.A.); (N.B.S.)
- Department of Nursing, Universidad Santiago de Cali, Cali 760001, Colombia
| | - Narly Benachi Sandoval
- Research Group “Health Care (Recognized by Colciencias)”, Universidad Santiago de Cali, Cali 760001, Colombia; (L.A.M.-U.); (D.V.A.); (N.B.S.)
- CAP Casanova, Consorci d’Atenció Primària de Salut Barcelona Esquerra, 08036 Barcelona, Spain
| | - Mauricio Hernández-Carrillo
- Health Faculty, Universidad del Valle, Cali 760001, Colombia;
- Health Faculty, Escuela Nacional del Deporte, Cali 760001, Colombia
| | - Guillermo Molina-Recio
- Department of Nursing, Pharmacology and Physiotherapy, University of Córdoba, 14014 Córdoba, Spain;
- Lifestyles, Innovation and Health (GA-16), Maimonides Biomedical Research Institute of Córdoba (IMIBIC), 14014 Córdoba, Spain
| |
Collapse
|
12
|
Gazzaroli G, Angeli A, Giacomini A, Ronca R. Proteasome inhibitors as anticancer agents. Expert Opin Ther Pat 2023; 33:775-796. [PMID: 37847492 DOI: 10.1080/13543776.2023.2272648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/16/2023] [Indexed: 10/18/2023]
Abstract
INTRODUCTION The therapeutic targeting of the ubiquitin-proteasome pathway (UPP) through inhibitors of the 20S proteasome core proteolytic activities has revolutionized the treatment of hematological malignancies and is paving the way for its extension to solid tumors. AREAS COVERED This review covers the progress made in the field of proteasome inhibitors, ranging from the first-generation bortezomib to the latest second-generation inhibitors such as carfilzomib and ixazomib as well as the proteasome inhibitors in clinical phase such as oprozomib and marizomib. The development of selective and potent proteasome inhibitors with improved pharmacological properties is described from the synthesis to their basic biological, and clinical validation. EXPERT OPINION Proteasome inhibitors have transformed the treatment landscape for hematological malignancies and hold great promise for cancer therapy. Combination therapies targeting multiple pathways, the development of novel inhibitors or 'hybrid-inhibitors,' and the optimization of treatment protocols are key areas for future exploration. The extension of proteasome inhibitors for the treatment of solid tumors, and their ability to pass the blood-brain barrier open new possibilities for treating central nervous system cancers. However, managing adverse effects, particularly those affecting the central nervous system, remains a critical consideration and a strategic 'working on' aspect for the near future.
Collapse
Affiliation(s)
- Giorgia Gazzaroli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Andrea Angeli
- Neurofarba Department, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
13
|
Abdalbari FH, Martinez-Jaramillo E, Forgie BN, Tran E, Zorychta E, Goyeneche AA, Sabri S, Telleria CM. Auranofin Induces Lethality Driven by Reactive Oxygen Species in High-Grade Serous Ovarian Cancer Cells. Cancers (Basel) 2023; 15:5136. [PMID: 37958311 PMCID: PMC10650616 DOI: 10.3390/cancers15215136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
High-grade serous ovarian cancer (HGSOC) accounts for 70% of ovarian cancer cases, and the survival rate remains remarkably low due to the lack of effective long-term consolidation therapies. Clinical remission can be temporarily induced by platinum-based chemotherapy, but death subsequently results from the extensive growth of a platinum-resistant component of the tumor. This work explores a novel treatment against HGSOC using the gold complex auranofin (AF). AF primarily functions as a pro-oxidant by inhibiting thioredoxin reductase (TrxR), an antioxidant enzyme overexpressed in ovarian cancer. We investigated the effect of AF on TrxR activity and the various mechanisms of cytotoxicity using HGSOC cells that are clinically sensitive or resistant to platinum. In addition, we studied the interaction between AF and another pro-oxidant, L-buthionine sulfoximine (L-BSO), an anti-glutathione (GSH) compound. We demonstrated that AF potently inhibited TrxR activity and reduced the vitality and viability of HGSOC cells regardless of their sensitivities to platinum. We showed that AF induces the accumulation of reactive oxygen species (ROS), triggers the depolarization of the mitochondrial membrane, and kills HGSOC cells by inducing apoptosis. Notably, AF-induced cell death was abrogated by the ROS-scavenger N-acetyl cysteine (NAC). In addition, the lethality of AF was associated with the activation of caspases-3/7 and the generation of DNA damage, effects that were also prevented by the presence of NAC. Finally, when AF and L-BSO were combined, we observed synergistic lethality against HGSOC cells, which was mediated by a further increase in ROS and a decrease in the levels of the antioxidant GSH. In summary, our results support the concept that AF can be used alone or in combination with L-BSO to kill HGSOC cells regardless of their sensitivity to platinum, suggesting that the depletion of antioxidants is an efficient strategy to mitigate the course of this disease.
Collapse
Affiliation(s)
- Farah H. Abdalbari
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 2B4, Canada; (F.H.A.); (E.M.-J.); (B.N.F.); (E.T.); (E.Z.); (A.A.G.)
| | - Elvis Martinez-Jaramillo
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 2B4, Canada; (F.H.A.); (E.M.-J.); (B.N.F.); (E.T.); (E.Z.); (A.A.G.)
| | - Benjamin N. Forgie
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 2B4, Canada; (F.H.A.); (E.M.-J.); (B.N.F.); (E.T.); (E.Z.); (A.A.G.)
| | - Estelle Tran
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 2B4, Canada; (F.H.A.); (E.M.-J.); (B.N.F.); (E.T.); (E.Z.); (A.A.G.)
| | - Edith Zorychta
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 2B4, Canada; (F.H.A.); (E.M.-J.); (B.N.F.); (E.T.); (E.Z.); (A.A.G.)
| | - Alicia A. Goyeneche
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 2B4, Canada; (F.H.A.); (E.M.-J.); (B.N.F.); (E.T.); (E.Z.); (A.A.G.)
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
| | - Siham Sabri
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
| | - Carlos M. Telleria
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 2B4, Canada; (F.H.A.); (E.M.-J.); (B.N.F.); (E.T.); (E.Z.); (A.A.G.)
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
| |
Collapse
|
14
|
Choudhary D, Kaur A, Singh P, Chaudhary G, Kaur R, Bayan MF, Chandrasekaran B, Marji SM, Ayman R. Target protein degradation by protacs: A budding cancer treatment strategy. Pharmacol Ther 2023; 250:108525. [PMID: 37696366 DOI: 10.1016/j.pharmthera.2023.108525] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023]
Abstract
Cancer is one of the most common causes of death. So, its lethal effect increases with time. Near about hundreds of cancers are known in humans. Cancer treatment is done to cure or prolonged remission, and shrinkage of the tumor. Cytotoxic agents, biological agents/targeted drugs, hormonal drugs, surgery, radiotherapy/proton therapy, chemotherapy, immunotherapy, and gene therapy are currently used in the treatment of cancer but their cost is high and cause various side effects. Seeing this, some new targeted strategies such as PROTACs are the need of the time. Proteolysis targeting chimera (PROTAC) has become one of the most discussed topics regarding cancer treatment. Few of the PROTAC molecules are in the trial phases. PROTACs have many advantages over other strategies such as modularity, compatibility, sub-stoichiometric activity, acting on undruggable targets, molecular design, and acts on intracellular targets, selectivity and specificity can be recruited for any cancer, versatility, and others. PROTACs are having some unclear questions on their pharmacokinetics, heavy-molecular weight, etc. PROTACs are anticipated to bring about a conversion in current healthcare and will emerge as booming treatments. In this review article we summarize PROTACs, their mechanism of action, uses, advantages, disadvantages, challenges, and future aspects for the successful development of potent PROTACs as a drug strategy.
Collapse
Affiliation(s)
- Diksha Choudhary
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Amritpal Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Pargat Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Gaurav Chaudhary
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India.
| | - Mohammad F Bayan
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| | | | - Saeed M Marji
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| | - Reema Ayman
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| |
Collapse
|
15
|
Monittola F, Bianchi M, Nasoni MG, Luchetti F, Magnani M, Crinelli R. Gastric cancer cell types display distinct proteasome/immunoproteasome patterns associated with migration and resistance to proteasome inhibitors. J Cancer Res Clin Oncol 2023; 149:10085-10097. [PMID: 37261527 PMCID: PMC10423134 DOI: 10.1007/s00432-023-04948-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/26/2023] [Indexed: 06/02/2023]
Abstract
PURPOSE Gastric cancers (GC) display histological and molecular differences. This heterogeneity has limited the development of new therapeutic strategies which requires the identification of the molecular players involved in GC pathogenesis and the investigation of their responsiveness to drugs. Several proteasome subunits have been identified as prognostic markers in GC and their role studied by gene knockdown. However, proteasomes are multi-subunit protein complexes co-existing in multiple forms with distinct activity/specificity and ability to change in response to inhibitors. Information on the role of different proteasome particles in cancer and their relevance as therapeutic targets is limited. METHODS Based on this evidence, subunit assembly into proteasome complexes and activity were investigated by native PAGE followed by immunoblotting, and by using fluorogenic substrates, respectively. RESULTS Here we show that GC cell lines with epithelial and/or diffuse Lauren's histotype express different levels of immunoproteasome subunits and equal amounts of constitutive counterparts. Immunoproteasome subunits were highly expressed and preferentially assembled into 19S capped complexes in diffuse-type cells, where most of the activity was catalyzed by the 26S and 30S particles. In epithelial cells, activity appeared equally distributed between 19S- and 11S-capped proteolytic particles. This proteasome pattern was associated with higher resistance of diffuse-type cells to proteasome inhibition. Immunoproteasome inhibition by ONX 0914 did not influence cell viability but affected metastatic cell migration. CONCLUSIONS These results suggest that pharmacological inhibition of the immunoproteasome may be useful in treating metastatic gastric cancers.
Collapse
Affiliation(s)
- Francesca Monittola
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| | - Marzia Bianchi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| | - Maria Gemma Nasoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| | - Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy
| | - Rita Crinelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029, Urbino, PU, Italy.
| |
Collapse
|
16
|
Kim Y, Kim EK, Chey Y, Song MJ, Jang HH. Targeted Protein Degradation: Principles and Applications of the Proteasome. Cells 2023; 12:1846. [PMID: 37508510 PMCID: PMC10378610 DOI: 10.3390/cells12141846] [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: 06/05/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
The proteasome is a multi-catalytic protease complex that is involved in protein quality control via three proteolytic activities (i.e., caspase-, trypsin-, and chymotrypsin-like activities). Most cellular proteins are selectively degraded by the proteasome via ubiquitination. Moreover, the ubiquitin-proteasome system is a critical process for maintaining protein homeostasis. Here, we briefly summarize the structure of the proteasome, its regulatory mechanisms, proteins that regulate proteasome activity, and alterations to proteasome activity found in diverse diseases, chemoresistant cells, and cancer stem cells. Finally, we describe potential therapeutic modalities that use the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Yosup Kim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Eun-Kyung Kim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Yoona Chey
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Min-Jeong Song
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Ho Hee Jang
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
17
|
Vottonen L, Koskela A, Felszeghy S, Wylegala A, Kryszan K, Gurubaran IS, Kaarniranta K, Wylegala E. Oxidative Stress and Cellular Protein Accumulation Are Present in Keratoconus, Macular Corneal Dystrophy, and Fuchs Endothelial Corneal Dystrophy. J Clin Med 2023; 12:4332. [PMID: 37445366 DOI: 10.3390/jcm12134332] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of the study was to investigate oxidative stress as well as cellular protein accumulation in corneal diseases including keratoconus (KC), macular corneal dystrophy (MCD), and Fuchs endothelial corneal dystrophy (FECD) at their primary affecting sites. Corneal buttons from KC, MCD, and FECD patients, as well as healthy controls, were analyzed immunohistochemically to evaluate the presence of oxidative stress and the function of the proteostasis network. 4-Fydroxynonenal (4-HNE) was used as a marker of oxidative stress, whereas the levels of catalase and heat-shock protein 70 (HSP70) were analyzed to evaluate the response of the antioxidant defense system and molecular chaperones, respectively. Sequestosome 1 (SQSTM1) levels were determined to assess protein aggregation and the functionality of autophagic degradation. Basal epithelial cells of the KC samples showed increased levels of oxidative stress marker 4-HNE and antioxidant enzyme catalase together with elevated levels of HSP70 and accumulation of SQSTM1. Corneal stromal cells and endothelial cells from MCD and FECD samples, respectively, showed similarly increased levels of these markers. All corneal diseases showed the presence of oxidative stress and activation of the molecular chaperone response to sustain protein homeostasis. However, the accumulation of protein aggregates suggests insufficient function of the protective mechanisms to limit the oxidative damage and removal of protein aggregates via autophagy. These results suggest that oxidative stress has a role in KC, MCD, and FECD at the cellular level as a secondary outcome. Thus, antioxidant- and autophagy-targeted therapies could be included as supporting care when treating KC or corneal dystrophies.
Collapse
Affiliation(s)
- Linda Vottonen
- Department of Ophthalmology, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Ali Koskela
- Department of Ophthalmology, University of Eastern Finland, 70210 Kuopio, Finland
| | - Szabolcs Felszeghy
- Institute of Biomedicine, University of Eastern Finland, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Adam Wylegala
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Faculty of Medical Sciences, Medical University of Silesia, 40-055 Katowice, Poland
- Ophthalmology Department, Railway Hospital, 40-760 Katowice, Poland
| | | | | | - Kai Kaarniranta
- Department of Ophthalmology, Kuopio University Hospital, 70210 Kuopio, Finland
- Department of Ophthalmology, University of Eastern Finland, 70210 Kuopio, Finland
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Edward Wylegala
- Ophthalmology Department, Railway Hospital, 40-760 Katowice, Poland
- Clinical Department of Ophthalmology, II School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, 40-760 Katowice, Poland
| |
Collapse
|
18
|
Zhang X, Nadolny C, Chen Q, Ali W, Hashmi SF, Deng R. Dysregulation and oncogenic activities of ubiquitin specific peptidase 2a in the pathogenesis of hepatocellular carcinoma. Am J Cancer Res 2023; 13:2392-2409. [PMID: 37424823 PMCID: PMC10326592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/09/2023] [Indexed: 07/11/2023] Open
Abstract
Ubiquitin specific peptidase 2a (USP2a) plays critical roles in protein degradation and other cellular activities. Currently, our understanding on USP2a dysregulation in subjects with hepatocellular carcinoma (HCC) and its roles in HCC pathogenesis is limited. In this study, we found that USP2a mRNA and protein levels were significantly upregulated in HCC tumors from both human and mice. USP2a overexpression in HepG2 and Huh 7 cells significantly increased cell proliferation while inhibition of USP2a activity by chemical inhibitor or stable knockout of USP2 by CRISPR markedly reduced cell proliferation. In addition, USP2a overexpression significantly augmented the resistance while knockout of USP2a markedly increased the susceptibility of HepG2 cells to bile acid-induced apoptosis and necrosis. Consistent with the oncogenic activities detected in vitro, overexpression of USP2a promoted de novo HCC development in mice with significantly increased tumor occurrence rates, tumor sizes and liver/body ratios. Further investigations with unbiased co-immunoprecipitation (Co-IP)-coupled proteomic analysis and Western blot identified novel USP2a target proteins involved in cell proliferation, apoptosis, and tumorigenesis. Analysis of those USP2a target proteins revealed that USP2a's oncogenic activities are mediated through multiple pathways, including modulating protein folding and assembling through regulating protein chaperones/co-chaperones HSPA1A, DNAJA1 and TCP1, promoting DNA replication and transcription through regulating RUVBL1, PCNA and TARDBP, and altering mitochondrial apoptotic pathway through regulating VDAC2. Indeed, those newly identified USP2a target proteins were markedly dysregulated in HCC tumors. In summary, USP2a was upregulated in HCC subjects and acted as an oncogene in the pathogenesis of HCC through multiple downstream pathways. The findings provided molecular and pathogenesis bases for developing interventions to treat HCC by targeting USP2a or its downstream pathways.
Collapse
Affiliation(s)
- Xinmu Zhang
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island 7 Greenhouse Road, Kingston, RI 02881, USA
| | - Christina Nadolny
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island 7 Greenhouse Road, Kingston, RI 02881, USA
| | - Qiwen Chen
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island 7 Greenhouse Road, Kingston, RI 02881, USA
| | - Winifer Ali
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island 7 Greenhouse Road, Kingston, RI 02881, USA
| | - Syed F Hashmi
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island 7 Greenhouse Road, Kingston, RI 02881, USA
| | - Ruitang Deng
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island 7 Greenhouse Road, Kingston, RI 02881, USA
| |
Collapse
|
19
|
Wong KC, Jayapalan JJ, Subramanian P, Ismail MN, Abdul-Rahman PS. Label-free quantitative mass spectrometry analysis of the circadian proteome of Drosophila melanogaster lethal giant larvae mutants reveals potential therapeutic effects of melatonin. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2023; 113:e22008. [PMID: 36915983 DOI: 10.1002/arch.22008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/10/2023] [Accepted: 02/22/2023] [Indexed: 05/16/2023]
Abstract
Mutation in the Drosophila melanogaster lethal giant larvae (lgl), a tumor suppressor gene with a well-established role in cellular polarity, is known to results in massive cellular proliferation and neoplastic outgrowths. Although the tumorigenic properties of lgl mutant have been previously studied, however, little is known about its consequences on the proteome. In this study, mass spectrometry-based label-free quantitative proteomics was employed to investigate the changes in the head and intestinal tissues proteins of Drosophila melanogaster, due to lgl mutation and following treatment with melatonin. Additionally, to uncover the time-influenced variations in the proteome during tumorigenesis and melatonin treatment, the rhythmic expression of proteins was also investigated at 6-h intervals within 24-h clock. Together, the present study has identified 434 proteins of altered expressions (p < 0.05 and fold change ±1.5) in the tissues of flies in response to lgl mutation as well as posttreatment with melatonin. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of differentially expressed proteins revealed that lgl mutation had significantly affected the biological functions, including metabolism, and protein synthesis and degradation, in flies' tissues. Besides, melatonin had beneficially mitigated the deleterious effects of lgl mutation by reversing the alterations in protein expression closer to baseline levels. Further, changes in protein expression in the tissues due to lgl mutation and melatonin treatment were found rhythmically orchestrated. Together, these findings provide novel insight into the pathways involved in lgl-induced tumorigenesis as well as demonstrated the efficacy of melatonin as a potential anticancer agent. Data are available via ProteomeXchange with identifier PXD033191.
Collapse
Affiliation(s)
- Kar-Cheng Wong
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Jaime J Jayapalan
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Universiti Malaya Centre for Proteomics Research (UMCPR), Universiti Malaya, Kuala Lumpur, Malaysia
| | - Perumal Subramanian
- Department of Biochemistry and Biotechnology, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Mohd Nazri Ismail
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, Bayan Lepas, Penang, Malaysia
| | - Puteri S Abdul-Rahman
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Universiti Malaya Centre for Proteomics Research (UMCPR), Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
20
|
Du T, Song Y, Ray A, Wan X, Yao Y, Samur MK, Shen C, Penailillo J, Sewastianik T, Tai YT, Fulciniti M, Munshi NC, Wu H, Carrasco RD, Chauhan D, Anderson KC. Ubiquitin receptor PSMD4/Rpn10 is a novel therapeutic target in multiple myeloma. Blood 2023; 141:2599-2614. [PMID: 36630605 PMCID: PMC10273170 DOI: 10.1182/blood.2022017897] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/12/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023] Open
Abstract
PSMD4/Rpn10 is a subunit of the 19S proteasome unit that is involved with feeding target proteins into the catalytic machinery of the 26S proteasome. Because proteasome inhibition is a common therapeutic strategy in multiple myeloma (MM), we investigated Rpn10 and found that it is highly expressed in MM cells compared with normal plasma cells. Rpn10 levels inversely correlated with overall survival in patients with MM. Inducible knockout or knockdown of Rpn10 decreased MM cell viability both in vitro and in vivo by triggering the accumulation of polyubiquitinated proteins, cell cycle arrest, and apoptosis associated with the activation of caspases and unfolded protein response-related pathways. Proteomic analysis revealed that inhibiting Rpn10 increased autophagy, antigen presentation, and the activation of CD4+ T and natural killer cells. We developed an in vitro AlphaScreen binding assay for high-throughput screening and identified a novel Rpn10 inhibitor, SB699551 (SB). Treating MM cell lines, leukemic cell lines, and primary cells from patients with MM with SB decreased cell viability without affecting the viability of normal peripheral blood mononuclear cells. SB inhibited the proliferation of MM cells even in the presence of the tumor-promoting bone marrow milieu and overcame proteasome inhibitor (PI) resistance without blocking the 20S proteasome catalytic function or the 19S deubiquitinating activity. Rpn10 blockade by SB triggered MM cell death via similar pathways as the genetic strategy. In MM xenograft models, SB was well tolerated, inhibited tumor growth, and prolonged survival. Our data suggest that inhibiting Rpn10 will enhance cytotoxicity and overcome PI resistance in MM, providing the basis for further optimization studies of Rpn10 inhibitors for clinical application.
Collapse
Affiliation(s)
- Ting Du
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Yan Song
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Arghya Ray
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Xueping Wan
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Yao Yao
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Mehmet K. Samur
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
- Department of Biostatistics and Computational Biology, Harvard T.H. Chan School of Public Health, Harvard Medical School, Harvard University, Boston, MA
| | - Chen Shen
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Harvard University, Boston, MA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA
| | - Johany Penailillo
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Tomasz Sewastianik
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Yu-Tzu Tai
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Mariateresa Fulciniti
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Nikhil C. Munshi
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
- VA Boston Healthcare System, Boston, MA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Harvard University, Boston, MA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA
| | - Ruben D. Carrasco
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA
| | - Dharminder Chauhan
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Kenneth C. Anderson
- Department of Medical Oncology, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| |
Collapse
|
21
|
Xu W, Graves A, Weisz-Hubshman M, Hegazy L, Magyar C, Liu Z, Nasiotis E, Samee MAH, Burris T, Lalani S, Zhang L. Assigning pathogenicity for TAB2 variants using a novel scalable functional assay and expanding TAB2 disease spectrum. Hum Mol Genet 2023; 32:959-970. [PMID: 36229919 PMCID: PMC9990987 DOI: 10.1093/hmg/ddac252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/13/2022] Open
Abstract
Haploinsufficiency of TGF-beta-activated kinase 1 (MAP3K7) binding protein 2 (TAB2) has been associated with congenital heart disease and more recently multiorgan structural abnormalities. Missense variant represents a major proportion of non-synonymous TAB2 variants reported in gnomAD (295/576) and Clinvar (16/73), most of which are variants of uncertain significance (VUSs). However, interpretation of TAB2 missense variants remains challenging because of lack of functional assays. To address this issue, we established a cell-based luciferase assay that enables high-throughput screening of TAB2 variants to assess the functional consequence for predicting variant pathogenicity. Using this platform, we screened 47 TAB2 variants including five pathogenic controls and one benign control, and the results showed that the transcriptional activity of activator protein 1 (AP-1) but not nuclear factor kappa B predicts the TAB2 variant pathogenicity. This assay provides accurate functional readout for both loss-of-function (LOF) and gain-of-function variants, which are associated with distinct phenotypes. In all, 22 out of 32 tested VUSs were reclassified. Genotype-Phenotype association showed that most patients with partial LOF variants do not exhibit congenital heart disease but high frequency of developmental delay, hypotonia and dysmorphic features, which suggests that genetic testing for TAB2 is needed for a broader spectrum of patients with more diverse phenotypes. Molecular modeling with Npl4 zinc finger (NZF) domain variants revealed that the stability of the NZF domain in TAB2 protein is crucial for AP-1 activation. In conclusion, we developed a highly effective functional assay for TAB2 variant prediction and interpretation.
Collapse
Affiliation(s)
- Weiyi Xu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Andrea Graves
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Monika Weisz-Hubshman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Lamees Hegazy
- Center for Clinical Pharmacology, Washington University School of Medicine and University of Health Sciences & Pharmacy, St. Louis, MO, USA
| | - Christina Magyar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Zian Liu
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Eleni Nasiotis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Md Abul Hassan Samee
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Thomas Burris
- Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Seema Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
22
|
MAPK4 silencing in gastric cancer drives liver metastasis by positive feedback between cancer cells and macrophages. Exp Mol Med 2023; 55:457-469. [PMID: 36797541 PMCID: PMC9981715 DOI: 10.1038/s12276-023-00946-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/08/2022] [Accepted: 12/13/2022] [Indexed: 02/18/2023] Open
Abstract
Liver metastasis is a major cause of death in gastric cancer patients, but the underlying mechanisms are poorly understood. Through a combination of in vivo screening and transcriptome profiling followed by quantitative RT-PCR and tissue array analyses, we found that mitogen-activated protein kinase 4 (MAPK4) downregulation in gastric cancer tissues from patients is significantly associated with liver metastasis and poor prognosis. The knockdown of MAPK4 in gastric cancer cells promotes liver metastasis in orthotopic mouse models. MAPK4 depletion in gastric cancer cells induces the secretion of macrophage migration inhibitory factor (MIF) to polarize tumor-associated macrophages (TAMs) in orthotopic xenograft tumors. Moreover, TAMs activate epithelial-mesenchymal transition of gastric cancer cells to suppress MAPK4 expression, which further increases MIF secretion to polarize TAMs. Taken together, our results suggest a previously undescribed positive feedback loop between cancer cells and macrophages mediated by MAPK4 silencing that facilitates gastric cancer liver metastasis.
Collapse
|
23
|
Hwangbo DS, Kwon YJ, Iwanaszko M, Jiang P, Abbasi L, Wright N, Alli S, Hutchison AL, Dinner AR, Braun RI, Allada R. Dietary Restriction Impacts Peripheral Circadian Clock Output Important for Longevity in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.04.522718. [PMID: 36711760 PMCID: PMC9881908 DOI: 10.1101/2023.01.04.522718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Circadian clocks may mediate lifespan extension by caloric or dietary restriction (DR). We find that the core clock transcription factor Clock is crucial for a robust longevity and fecundity response to DR in Drosophila. To identify clock-controlled mediators, we performed RNA-sequencing from abdominal fat bodies across the 24 h day after just 5 days under control or DR diets. In contrast to more chronic DR regimens, we did not detect significant changes in the rhythmic expression of core clock genes. Yet we discovered that DR induced de novo rhythmicity or increased expression of rhythmic clock output genes. Network analysis revealed that DR increased network connectivity in one module comprised of genes encoding proteasome subunits. Adult, fat body specific RNAi knockdown demonstrated that proteasome subunits contribute to DR-mediated lifespan extension. Thus, clock control of output links DR-mediated changes in rhythmic transcription to lifespan extension.
Collapse
Affiliation(s)
- Dae-Sung Hwangbo
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
- Department of Biology, University of Louisville, Louisville, 40292, KY, USA
| | - Yong-Jae Kwon
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Marta Iwanaszko
- Biostatistics Division, Department of Preventive Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
| | - Peng Jiang
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Ladan Abbasi
- Department of Biology, University of Louisville, Louisville, 40292, KY, USA
| | - Nicholas Wright
- Department of Biology, University of Louisville, Louisville, 40292, KY, USA
| | - Sarayu Alli
- Department of Biology, University of Louisville, Louisville, 40292, KY, USA
| | - Alan L. Hutchison
- James Franck Institute, Department of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Aaron R. Dinner
- James Franck Institute, Department of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Rosemary I Braun
- Biostatistics Division, Department of Preventive Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
| | - Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
24
|
Small-molecule inhibitors of proteasome increase CjCas9 protein stability. PLoS One 2023; 18:e0280353. [PMID: 36656806 PMCID: PMC9851528 DOI: 10.1371/journal.pone.0280353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/27/2022] [Indexed: 01/20/2023] Open
Abstract
The small size of CjCas9 can make easier its vectorization for in vivo gene therapy. However, compared to the SpCas9, the CjCas9 is, in general, less efficient to generate indels in target genes. The factors that affect its efficacity are not yet determined. We observed that the CjCas9 protein expressed in HEK293T cells after transfection of this transgene under a CMV promoter was much lower than the SpCas9 protein in the same conditions. We thus evaluated the effect of proteasome inhibitors on CjCas9 protein stability and its efficiency on FXN gene editing. Western blotting showed that the addition of MG132 or bortezomib, significantly increased CjCas9 protein levels in HEK293T and HeLa cells. Moreover, bortezomib increased the level of CjCas9 protein expressed under promoters weaker than CMV such as CBH or EFS but which are specific for certain tissues. Finally, ddPCR quantification showed that bortezomib treatment enhanced CjCas9 efficiency to delete GAA repeat region of FXN gene in HEK293T cells. The improvement of CjCas9 protein stability would facilitate its used in CRISPR/Cas system.
Collapse
|
25
|
Zinc and Zinc Transporters in Dermatology. Int J Mol Sci 2022; 23:ijms232416165. [PMID: 36555806 PMCID: PMC9785331 DOI: 10.3390/ijms232416165] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Zinc is an important trace mineral in the human body and a daily intake of zinc is required to maintain a healthy status. Over the past decades, zinc has been used in formulating topical and systemic therapies for various skin disorders owing to its wound healing and antimicrobial properties. Zinc transporters play a major role in maintaining the integrity of the integumentary system by controlling zinc homeostasis within dermal layers. Mutations and abnormal function of zinc-transporting proteins can lead to disease development, such as spondylocheirodysplastic Ehlers-Danlos syndrome (SCD-EDS) and acrodermatitis enteropathica (AE) which can be fatal if left untreated. This review discusses the layers of the skin, the importance of zinc and zinc transporters in each layer, and the various skin disorders caused by zinc deficiency, in addition to zinc-containing compounds used for treating different skin disorders and skin protection.
Collapse
|
26
|
Li J, Kou Y, Zhang X, Xiao X, Ou Y, Cao L, Guo M, Qi C, Wang Z, Liu Y, Shuai Q, Wang H, Yang S. Biochanin A inhibits lung adenocarcinoma progression by targeting ZEB1. Discov Oncol 2022; 13:138. [PMID: 36512117 PMCID: PMC9748019 DOI: 10.1007/s12672-022-00601-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Lung adenocarcinoma is the major subtype of lung cancer, accounting for approximately 40% of lung cancers. During clinical treatment, the emergence of chemotherapy resistance seriously affects the effectiveness of treatment. Thus, finding new chemotherapeutic sensitizers is considered to be one of the effective solutions. Biochanin A, as a naturally occurring isoflavone, has been demonstrated to exhibit anticancer effects in various tumors. However, the potential mechanisms of Biochanin A to inhibit tumor development have not been clarified. In the present study, we found that the combinational treatment of cisplatin and Biochanin A exhibited strong synergistic repression on lung adenocarcinoma growth and progression in vitro and in vivo. Considering that epithelial-mesenchymal transition (EMT) is recognized to be associated with both chemoresistance and metastasis, we examined the EMT-related markers and found that Biochanin A could specifically inhibit the expression of ZEB1. Importantly, Biochanin A chemosensitizes lung adenocarcinoma and inhibits cancer cell metastasis by suppressing ZEB1. At the molecular level, Biochanin A affects the stability of ZEB1 protein through the deubiquitination pathway and thereby influences the progression of lung adenocarcinoma. In conclusion, our finding elucidates the potential efficacy of Bichanin A as a chemosensitizer and provides new strategy for the chemotherapy of advanced lung adenocarcinoma.
Collapse
Affiliation(s)
- Jianjun Li
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, 215006, Suzhou, China
| | - Yaqi Kou
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China
| | - Xiaohan Zhang
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China
| | - Xuechun Xiao
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China
| | - Yang Ou
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China
| | - Lixia Cao
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China
| | - Min Guo
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China
| | - Chunchun Qi
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China
| | - Zhaoyang Wang
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China
| | - Yuxin Liu
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China
| | - Qiuying Shuai
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China
| | - Hang Wang
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China.
- Medical College of Nankai University, 94 Weijin Road, 300071, Tianjin, China.
| | - Shuang Yang
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, 300071, Tianjin, China.
- Institute of Transplantation Medicine, Nankai University, 300071, Tianjin, China.
- Medical College of Nankai University, 94 Weijin Road, 300071, Tianjin, China.
| |
Collapse
|
27
|
USP7 Inhibitors in Cancer Immunotherapy: Current Status and Perspective. Cancers (Basel) 2022; 14:cancers14225539. [PMID: 36428632 PMCID: PMC9688046 DOI: 10.3390/cancers14225539] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 11/12/2022] Open
Abstract
Ubiquitin-specific protease 7 (USP7) regulates the stability of a plethora of intracellular proteins involved in the suppression of anti-tumor immune responses and its overexpression is associated with poor survival in many cancers. USP7 impairs the balance of the p53/MDM2 axis resulting in the proteasomal degradation of the p53 tumor suppressor, a process that can be reversed by small-molecule inhibitors of USP7. USP7 was shown to regulate the anti-tumor immune responses in several cases. Its inhibition impedes the function of regulatory T cells, promotes polarization of tumor-associated macrophages, and reduces programmed death-ligand 1 (PD-L1) expression in tumor cells. The efficacy of small-molecule USP7 inhibitors was demonstrated in vivo. The synergistic effect of combining USP7 inhibition with cancer immunotherapy is a promising therapeutic approach, though its clinical efficacy is yet to be proven. In this review, we focus on the recent developments in understanding the intrinsic role of USP7, its interplay with other molecular pathways, and the therapeutic potential of targeting USP7 functions.
Collapse
|
28
|
Harju N. Regulation of oxidative stress and inflammatory responses in human retinal pigment epithelial cells. Acta Ophthalmol 2022; 100 Suppl 273:3-59. [DOI: 10.1111/aos.15275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Niina Harju
- School of Pharmacy University of Eastern Finland Kuopio Finland
| |
Collapse
|
29
|
Kwon JE, Jo SH, Song WS, Lee JS, Jeon HJ, Park JH, Kim YR, Baek JH, Kim MG, Kwon SY, Kim JS, Yang YH, Kim YG. Investigation of metabolic crosstalk between host and pathogenic Clostridioides difficile via multiomics approaches. Front Bioeng Biotechnol 2022; 10:971739. [PMID: 36118584 PMCID: PMC9478559 DOI: 10.3389/fbioe.2022.971739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
Clostridioides difficile is a gram-positive anaerobic bacterium that causes antibiotic-associated infections in the gut. C. difficile infection develops in the intestine of a host with an imbalance of the intestinal microbiota and, in severe cases, can lead to toxic megacolon, intestinal perforation, and even death. Despite its severity and importance, however, the lack of a model to understand host-pathogen interactions and the lack of research results on host cell effects and response mechanisms under C. difficile infection remain limited. Here, we developed an in vitro anaerobic-aerobic C. difficile infection model that enables direct interaction between human gut epithelial cells and C. difficile through the Mimetic Intestinal Host–Microbe Interaction Coculture System. Additionally, an integrative multiomics approach was applied to investigate the biological changes and response mechanisms of host cells caused by C. difficile in the early stage of infection. The C. difficile infection model was validated through the induction of disaggregation of the actin filaments and disruption of the intestinal epithelial barrier as the toxin-mediated phenotypes following infection progression. In addition, an upregulation of stress-induced chaperones and an increase in the ubiquitin proteasomal pathway were identified in response to protein stress that occurred in the early stage of infection, and downregulation of proteins contained in the electron transfer chain and ATP synthase was observed. It has been demonstrated that host cell energy metabolism is inhibited through the glycolysis of Caco-2 cells and the reduction of metabolites belonging to the TCA cycle. Taken together, our C. difficile infection model suggests a new biological response pathway in the host cell induced by C. difficile during the early stage of infection at the molecular level under anaerobic-aerobic conditions. Therefore, this study has the potential to be applied to the development of future therapeutics through basic metabolic studies of C. difficile infection.
Collapse
Affiliation(s)
- Ji-Eun Kwon
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Won-Suk Song
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Jae-Seung Lee
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Hyo-Jin Jeon
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Ji-Hyeon Park
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Ye-Rim Kim
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Ji-Hyun Baek
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Min-Gyu Kim
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Seo-Young Kwon
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Yung-Hun Yang
- Department of Biological Engineering, Konkuk University, Seoul, South Korea
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
- *Correspondence: Yun-Gon Kim,
| |
Collapse
|
30
|
Pei Y, Chen S, Zhang Y, Olga V, Li Y, Diao X, Zhou H. Coral and it's symbionts responses to the typical global marine pollutant BaP by 4D-Proteomics approach. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 307:119440. [PMID: 35623566 DOI: 10.1016/j.envpol.2022.119440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/13/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
The symbiosis of corals, zooxanthellae, and microbes is the foundation of the coral reef ecosystem. In addition to global warming, marine pollutants are another important factor causing the breakdown of coral symbiosis. Benzo(a)pyrene (BaP) is a globally widespread marine environmental pollutant that poses a severe threat to marine ecosystems. However, responses of coral symbionts to global marine pollutant stress remain unclear. In this study, we selected Acropora formosa as the target coral to explore its response to 50 μg L-1 BaP stress using diaPASEF proteomics and 16s rRNA microbiome analysis. The results showed that: 1) the coral symbionts were sensitive to BaP stress; 2) the photosynthetic system of zooxanthellae was crucial for the balance of symbiotic relationships; 3) the destruction of the photosynthetic system induced a zooxanthellae hypoxic stress response; 4) corals adapted to BaP stress by promoting non-essential protein degradation and changing energy metabolism strategies; 5) symbiotic bacteria showed strong adaptability to BaP. This study not only fills the gap in understanding the response mechanism of coral symbionts under BaP stress, but also provides fundamental data for coral reef protection strategies.
Collapse
Affiliation(s)
- Yuebin Pei
- State Key Laboratory of South China Sea Marine Resource Utilisation, Hainan University, Haikou, 570228, China; School of Life Sciences, Hainan University, Haikou, 570228, China; One Health Institute, Hainan University, Haikou, Hainan, 570228, China
| | - Shuai Chen
- State Key Laboratory of South China Sea Marine Resource Utilisation, Hainan University, Haikou, 570228, China; School of Life Sciences, Hainan University, Haikou, 570228, China; One Health Institute, Hainan University, Haikou, Hainan, 570228, China
| | - Yuting Zhang
- State Key Laboratory of South China Sea Marine Resource Utilisation, Hainan University, Haikou, 570228, China; School of Life Sciences, Hainan University, Haikou, 570228, China; One Health Institute, Hainan University, Haikou, Hainan, 570228, China
| | - Volovych Olga
- State Key Laboratory of South China Sea Marine Resource Utilisation, Hainan University, Haikou, 570228, China; School of Life Sciences, Hainan University, Haikou, 570228, China
| | - Yuanchao Li
- Hainan Academy of Ocean and Fisheries Sciences, Haikou, 571126, China
| | - Xiaoping Diao
- State Key Laboratory of South China Sea Marine Resource Utilisation, Hainan University, Haikou, 570228, China
| | - Hailong Zhou
- State Key Laboratory of South China Sea Marine Resource Utilisation, Hainan University, Haikou, 570228, China; School of Life Sciences, Hainan University, Haikou, 570228, China; One Health Institute, Hainan University, Haikou, Hainan, 570228, China.
| |
Collapse
|
31
|
Kim J, Park A, Hwang J, Zhao X, Kwak J, Kim HW, Ku M, Yang J, Kim TI, Jeong KS, Choi U, Lee H, Shin SJ. KS10076, a chelator for redox-active metal ions, induces ROS-mediated STAT3 degradation in autophagic cell death and eliminates ALDH1 + stem cells. Cell Rep 2022; 40:111077. [PMID: 35858554 DOI: 10.1016/j.celrep.2022.111077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 03/17/2022] [Accepted: 06/20/2022] [Indexed: 12/21/2022] Open
Abstract
Redox-active metal ions are pivotal for rapid metabolism, proliferation, and aggression across cancer types, and this presents metal chelation as an attractive cancer cell-targeting strategy. Here, we identify a metal chelator, KS10076, as a potent anti-cancer drug candidate. A metal-bound KS10076 complex with redox potential for generating hydrogen peroxide and superoxide anions induces intracellular reactive oxygen species (ROS). The elevation of ROS by KS10076 promotes the destabilization of signal transducer and activator of transcription 3, removes aldehyde dehydrogenase isoform 1-positive cancer stem cells, and subsequently induces autophagic cell death. Bioinformatic analysis of KS10076 susceptibility in pan-cancer cells shows that KS10076 potentially targets cancer cells with increased mitochondrial function. Furthermore, patient-derived organoid models demonstrate that KS10076 efficiently represses cancer cells with active KRAS, and fluorouracil resistance, which suggests clinical advantages.
Collapse
Affiliation(s)
- Jaehee Kim
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Areum Park
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Jieon Hwang
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Xianghua Zhao
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jaesung Kwak
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Hyun Woo Kim
- Chemical Data-Driven Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Minhee Ku
- Department of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Severance of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jaemoon Yang
- Department of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Severance of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Tae Il Kim
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kyu-Sung Jeong
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Uyeong Choi
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyuk Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea.
| | - Sang Joon Shin
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
32
|
Barbosa-Silva A, Magalhães M, Da Silva GF, Da Silva FAB, Carneiro FRG, Carels N. A Data Science Approach for the Identification of Molecular Signatures of Aggressive Cancers. Cancers (Basel) 2022; 14:2325. [PMID: 35565454 PMCID: PMC9103663 DOI: 10.3390/cancers14092325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/04/2022] [Accepted: 03/12/2022] [Indexed: 02/05/2023] Open
Abstract
The main hallmarks of cancer include sustaining proliferative signaling and resisting cell death. We analyzed the genes of the WNT pathway and seven cross-linked pathways that may explain the differences in aggressiveness among cancer types. We divided six cancer types (liver, lung, stomach, kidney, prostate, and thyroid) into classes of high (H) and low (L) aggressiveness considering the TCGA data, and their correlations between Shannon entropy and 5-year overall survival (OS). Then, we used principal component analysis (PCA), a random forest classifier (RFC), and protein-protein interactions (PPI) to find the genes that correlated with aggressiveness. Using PCA, we found GRB2, CTNNB1, SKP1, CSNK2A1, PRKDC, HDAC1, YWHAZ, YWHAB, and PSMD2. Except for PSMD2, the RFC analysis showed a different list, which was CAD, PSMD14, APH1A, PSMD2, SHC1, TMEFF2, PSMD11, H2AFZ, PSMB5, and NOTCH1. Both methods use different algorithmic approaches and have different purposes, which explains the discrepancy between the two gene lists. The key genes of aggressiveness found by PCA were those that maximized the separation of H and L classes according to its third component, which represented 19% of the total variance. By contrast, RFC classified whether the RNA-seq of a tumor sample was of the H or L type. Interestingly, PPIs showed that the genes of PCA and RFC lists were connected neighbors in the PPI signaling network of WNT and cross-linked pathways.
Collapse
Affiliation(s)
- Adriano Barbosa-Silva
- Center for Medical Statistics, Informatics and Intelligent Systems, Institute for Artificial Intelligence, Medical University of Vienna, 1090 Vienna, Austria
- Centre for Translational Bioinformatics, William Harvey Research Institute, Queen Mary University of London, London E14NS, UK
- ITTM S.A.-Information Technology for Translational Medicine, Esch-sur-Alzette, 4354 Luxembourg, Luxembourg
| | - Milena Magalhães
- Plataforma de Modelagem de Sistemas Biológicos, Center for Technology Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
| | - Gilberto Ferreira Da Silva
- Plataforma de Modelagem de Sistemas Biológicos, Center for Technology Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
| | - Fabricio Alves Barbosa Da Silva
- Laboratório de Modelagem Computacional de Sistemas Biológicos, Scientific Computing Program, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
| | - Flávia Raquel Gonçalves Carneiro
- Center for Technology Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231050, Brazil
| | - Nicolas Carels
- Plataforma de Modelagem de Sistemas Biológicos, Center for Technology Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040900, Brazil
| |
Collapse
|
33
|
Chen F, Chen L, Li Y, Sang H, Zhang C, Yuan S, Yang J. TRAF3 Positively Regulates Host Innate Immune Resistance to Influenza A Virus Infection. Front Cell Infect Microbiol 2022; 12:839625. [PMID: 35573779 PMCID: PMC9093644 DOI: 10.3389/fcimb.2022.839625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor necrosis factor receptor-associated factor 3 (TRAF3) is one of the intracellular adaptor proteins for the innate immune response, which is involved in signaling regulation in various cellular processes, including the immune responses defending against invading pathogens. However, the defense mechanism of TRAF3 against influenza virus infection remains elusive. In this study, we found that TRAF3 could positively regulate innate antiviral response. Overexpression of TRAF3 significantly enhanced virus-induced IRF3 activation, IFN-β production, and antiviral response, while TRAF3 knockdown promoted influenza A virus replication. Moreover, we clarified that inhibiting ubiquitinated degradation of TRAF3 was associated with anti-influenza effect, thereby facilitating antiviral immunity upon influenza A virus infection. We further demonstrated the key domains of TRAF3 involved in anti-influenza effect. Taken together, these results suggested that TRAF3 performs a vital role in host defense against influenza A virus infection by the type-I IFN signaling pathway. Our findings provide insights into the development of drugs to prevent TRAF3 degradation, which could be a novel therapeutic approach for treatment of influenza A virus infection.
Collapse
Affiliation(s)
- Fangzhao Chen
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Department of Pharmacy, Shenzhen Children’s Hospital, Shenzhen, China
| | - Liurong Chen
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yinyan Li
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Huiting Sang
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Chunyu Zhang
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuofeng Yuan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jie Yang
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
34
|
Chen D, Song D, Ma Y, Lu W, Qiu J, Wang Y. USP22 promotes pro‑inflammatory responses in Pseudomonas aeruginosa‑induced keratitis by targeting TRAF6. Mol Med Rep 2022; 25:149. [PMID: 35244191 DOI: 10.3892/mmr.2022.12665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/11/2020] [Indexed: 11/06/2022] Open
Abstract
Pseudomonas aeruginosa (PA)‑induced keratitis is characterized by inflammatory epithelial edema, stromal infiltration, corneal ulceration and can lead to vision loss. The present study aimed to study the effect of ubiquitin‑specific protease 22 (USP22) on PA‑induced keratitis. Using RT‑qPCR and western blotting, significantly increased expression of USP22 was identified in mouse corneas and cultured RAW264.7 cells following PA stimulation. In addition, the results of in vivo experiments, western blot assay and ELISA suggested that the silencing of USP22 attenuated disease progression, downregulated the NF‑κB pathway and suppressed the expression of pro‑inflammatory cytokines following PA stimulation. Notably, it was identified that the expression of tumor necrosis factor receptor‑associated factor 6 (TRAF6) was decreased by silencing of USP22 and USP22 was found to remove lysine 48‑linked poly‑ubiquitination chains from TRAF6 to stabilize TRAF6 expression and these effects were clearly aggravated following PA infection.
Collapse
Affiliation(s)
- Di Chen
- Department of Ophthalmology, Second Affiliated Hospital of Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, Shandong 271000, P.R. China
| | - Dawei Song
- Department of Ophthalmology, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Yibin Ma
- Department of Ophthalmology, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Weizhao Lu
- Department of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, Shandong 271000, P.R. China
| | - Jianfeng Qiu
- Department of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, Shandong 271000, P.R. China
| | - Yi Wang
- Department of Ophthalmology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
35
|
Global identification of phospho-dependent SCF substrates reveals a FBXO22 phosphodegron and an ERK-FBXO22-BAG3 axis in tumorigenesis. Cell Death Differ 2022; 29:1-13. [PMID: 34215846 PMCID: PMC8738747 DOI: 10.1038/s41418-021-00827-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
SKP1-CUL1-F-box (SCF) ubiquitin ligases play fundamental roles in cellular functions. Typically, substrate phosphorylation is required for SCF recognition and subsequent degradation. However, phospho-dependent substrates remain largely unidentified. Here, using quantitative phoshoproteome approach, we performed a system-wide investigation of phospho-dependent SCF substrates. This strategy identified diverse phospho-dependent candidates. Biochemical verification revealed a mechanism by which SCFFBXO22 recognizes the motif XXPpSPXPXX as a conserved phosphodegron to target substrates for destruction. We further demonstrated BAG3, a HSP70 co-chaperone, is a bona fide substrate of SCFFBXO22. FBXO22 mediates BAG3 ubiquitination and degradation that requires ERK-dependent BAG3 phosphorylation at S377. FBXO22 depletion or expression of a stable BAG3 S377A mutant promotes tumor growth via defects in apoptosis and cell cycle progression in vitro and in vivo. In conclusion, our study identified broad phosphorylation-dependent SCF substrates and demonstrated a phosphodegron recognized by FBXO22 and a novel ERK-FBXO22-BAG3 axis involved in tumorigenesis.
Collapse
|
36
|
Mattes MJ, Mattes JA, Groisberg R, Mattes MD. Therapy of Angiosarcoma with Thalidomide and Lenalidomide. Case Rep Oncol 2021; 14:1580-1585. [PMID: 34949999 PMCID: PMC8647127 DOI: 10.1159/000519970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 09/29/2021] [Indexed: 02/02/2023] Open
Abstract
Angiosarcoma is an uncommon malignancy with a poor prognosis. Systemic therapy options for patients with metastatic disease generally have limited effectiveness. In this case study, a 73-year-old male with metastatic angiosarcoma who previously declined chemotherapy and developed progressive disease after checkpoint inhibitor immunotherapy elected to try thalidomide based on 6 case reports describing its effectiveness. Thalidomide resulted in stable disease for 9 months, but due to severe neuropathy as a side effect, lenalidomide was then substituted for thalidomide. The patient continued to have stable disease on lenalidomide for an additional 16 months and ongoing. This is the first case study to report on effective treatment of angiosarcoma with lenalidomide. Further investigation of lenalidomide in the management of angiosarcoma is warranted.
Collapse
Affiliation(s)
- M Jules Mattes
- Center for Molecular Medicine and Immunology, Garden State Cancer Center, Belleville, New Jersey, USA
| | - Jeffrey A Mattes
- Psychopharmacology Research Association, Princeton, New Jersey, USA
| | - Roman Groisberg
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Malcolm D Mattes
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
37
|
Shi J, Xiong Z, Wang K, Yuan C, Huang Y, Xiao W, Meng X, Chen Z, Lv Q, Miao D, Liang H, Xu T, Xie K, Yang H, Zhang X. HIF2α promotes tumour growth in clear cell renal cell carcinoma by increasing the expression of NUDT1 to reduce oxidative stress. Clin Transl Med 2021; 11:e592. [PMID: 34841698 PMCID: PMC8567048 DOI: 10.1002/ctm2.592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The key role of hypoxia-inducible factor 2alpha (HIF2α) in the process of renal cancer has been confirmed. In the field of tumour research, oxidative stress is also considered to be an important influencing factor. However, the relationship and biological benefits of oxidative stress and HIF2α in ccRCC remain unclear. This research attempts to explore the effect of oxidative stress on the cancer-promoting effect of HIF2α in ccRCC and reveal its mechanism of action. METHODS The bioinformatics analysis for ccRCC is based on whole transcriptome sequencing and TCGA database. The detection of the expression level of related molecules is realised by western blot and PCR. The expression of Nucleoside diphosphate-linked moiety X-type motif 1 (NUDT1) was knocked down by lentiviral infection technology. The functional role of NUDT1 were further investigated by CCK8 assays, transwell assays and cell oxidative stress indicator detection. The exploration of related molecular mechanisms is realised by Luciferase assays and Chromatin immunoprecipitation (ChIP) assays. RESULTS Molecular screening based on knockdown HIF2α sequencing data and oxidative stress related data sets showed that NUDT1 is considered to be an important molecule for the interaction of HIF2α with oxidative stress. Subsequent experimental results showed that NUDT1 can cooperate with HIF2α to promote the progression of ccRCC. And this biological effect was found to be caused by the oxidative stress regulated by NUDT1. Mechanistically, HIF2α transcription activates the expression of NUDT1, thereby inhibiting oxidative stress and promoting the progression of ccRCC. CONCLUSIONS This research clarified a novel mechanism by which HIF2α stabilises sirtuin 3 (SIRT3) through direct transcriptional activation of NUDT1, thereby inhibiting oxidative stress to promote the development of ccRCC. It provided the possibility for the selection of new therapeutic targets for ccRCC and the study of combination medication regimens.
Collapse
Affiliation(s)
- Jian Shi
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Zhiyong Xiong
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Keshan Wang
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Changfei Yuan
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Yu Huang
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Wen Xiao
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Xiangui Meng
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Zhixian Chen
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Qingyang Lv
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Daojia Miao
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Huageng Liang
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Tianbo Xu
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Kairu Xie
- Department of Pathogenic BiologySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Hongmei Yang
- Department of Pathogenic BiologySchool of Basic MedicineHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Xiaoping Zhang
- Department of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
- Institute of UrologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiP. R. China
| |
Collapse
|
38
|
Sung K, Hosoya K, Murase Y, Deguchi T, Kim S, Sunaga T, Okumura M. Visualizing the cancer stem-like properties of canine tumour cells with low proteasome activity. Vet Comp Oncol 2021; 20:324-335. [PMID: 34719098 DOI: 10.1111/vco.12779] [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: 06/29/2021] [Accepted: 10/15/2021] [Indexed: 11/26/2022]
Abstract
Cancer stem-like cells (CSCs) cause treatment failure in various tumours; however, establishing CSC-targeted therapies has been hampered by difficulties in the identification and isolation of this small sub-population of cells. Recent studies have revealed that tumour cells with low proteasome activity display a CSC phenotype that can be utilized to image CSCs in canines. This study visualizes and reveals the CSC-like properties of tumour cells with low proteasome activity in HMPOS (osteosarcoma) and MegTCC (transitional cell carcinoma), which are canine cell lines. The parent cells were genetically engineered to express ZsGreen1, a fluorescent protein connected to the carboxyl-terminal degron of canine ornithine decarboxylase that accumulates with low proteasome activity (ZsG+ cells). ZsG+ cells were imaged and the mode of action of this system was confirmed using a proteasome inhibitor (MG-132), which increased the ZsGreen1 fluorescence intensity. The CSC-like properties of ZsG+ cells were evaluated on the basis of cell divisions, cell cycle, the expression of CSC markers and tumourigenicity. ZsG+ cells underwent asymmetric divisions and had a low percentage of G0/G1 phase cells; moreover, ZsG+ cells expressed CSC markers such as CD133 and showed a large tumourigenic capability. In histopathological analysis, ZsG+ cells were widely distributed in the tumour samples derived from ZsG+ cells and in the proliferative regions of the tumours. The results of this study indicate that visualized canine tumour cells with low proteasome activity have a CSC-like phenotype and that this visualization system can be utilized to identify and isolate canine CSCs.
Collapse
Affiliation(s)
- Koangyong Sung
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kenji Hosoya
- Veterinary Teaching Hospital, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yusuke Murase
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Tatsuya Deguchi
- Veterinary Teaching Hospital, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Sangho Kim
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Takafumi Sunaga
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Masahiro Okumura
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
39
|
Nadolny C, Zhang X, Chen Q, Hashmi SF, Ali W, Hemme C, Ahsan N, Chen Y, Deng R. Dysregulation and activities of ubiquitin specific peptidase 2b in the pathogenesis of hepatocellular carcinoma. Am J Cancer Res 2021; 11:4746-4767. [PMID: 34765291 PMCID: PMC8569343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/26/2021] [Indexed: 06/13/2023] Open
Abstract
Ubiquitin specific peptidase-2 (USP2) plays important roles in a myriad of cellular activities through deubiquitinating target proteins and its implications in various diseases, especially cancers, are starting to emerge. Our current understanding on USP2 expression in subjects with hepatocellular carcinoma (HCC) and its roles in the pathogenesis of HCC is limited. In this study, we found that USP2 protein and mRNA levels were significantly dysregulated in HCC tumor (HCC-T) when compared to adjacent non-tumor (HCC-NT) or normal liver tissues from both human and mouse HCC model. Among the USP2 isoforms, USP2b was the predominant isoform in the normal liver and markedly down-regulated in HCC-T tissues in both human and mice. Data from overexpression, chemical inhibition and knockout studies consistently demonstrated that USP2b promoted cell proliferation, colony formation and wound healing in HepG2 and Huh 7 cells. On the other hand, USP2b exhibited proapoptotic and pronecrtotic activities through enhancing bile acid-induced apoptosis and necrosis in both HepG2 and Huh 7 cells. Unbiased proteomic analysis of USP2-knockout (KO) and parental HepG2 cells resulted in identification of USP2-regulated downstream target proteins involved in cell proliferation, apoptosis, and tumorigenesis, including serine/threonine kinase 4 (STK4), epidermal growth factor receptor (EGFR), dipeptidyl peptidase 4 (DPP4) and fatty acid binding protein 1 (FABP1). In conclusion, USP2b expression was dysregulated in subjects with HCC and contributed to the pathogenesis of HCC by promoting cell proliferation and exerting proapoptotic and pronecrotic activities. The findings provide the molecular basis for developing therapies for HCC through modulating USP2b expression or activities.
Collapse
Affiliation(s)
- Christina Nadolny
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island7 Greenhouse Road, Kingston, RI 02881, USA
| | - Xinmu Zhang
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island7 Greenhouse Road, Kingston, RI 02881, USA
| | - Qiwen Chen
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island7 Greenhouse Road, Kingston, RI 02881, USA
| | - Syed F Hashmi
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island7 Greenhouse Road, Kingston, RI 02881, USA
| | - Winifer Ali
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island7 Greenhouse Road, Kingston, RI 02881, USA
| | - Christopher Hemme
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island7 Greenhouse Road, Kingston, RI 02881, USA
| | - Nagib Ahsan
- COBRE Center for Cancer Research Development, Proteomics Core Facility, Rhode Island HospitalProvidence, RI 02903, USA
- Division of Biology and Medicine, Brown UniversityProvidence, RI 02903, USA
- Department of Chemistry and Biochemistry, University of Oklahoma101 Stephenson Parkway, Norman, OK 73019, USA
| | - Yuan Chen
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island7 Greenhouse Road, Kingston, RI 02881, USA
| | - Ruitang Deng
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island7 Greenhouse Road, Kingston, RI 02881, USA
| |
Collapse
|
40
|
Gadhave K, Kumar D, Uversky VN, Giri R. A multitude of signaling pathways associated with Alzheimer's disease and their roles in AD pathogenesis and therapy. Med Res Rev 2021; 41:2689-2745. [PMID: 32783388 PMCID: PMC7876169 DOI: 10.1002/med.21719] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/13/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
The exact molecular mechanisms associated with Alzheimer's disease (AD) pathology continue to represent a mystery. In the past decades, comprehensive data were generated on the involvement of different signaling pathways in the AD pathogenesis. However, the utilization of signaling pathways as potential targets for the development of drugs against AD is rather limited due to the immense complexity of the brain and intricate molecular links between these pathways. Therefore, finding a correlation and cross-talk between these signaling pathways and establishing different therapeutic targets within and between those pathways are needed for better understanding of the biological events responsible for the AD-related neurodegeneration. For example, autophagy is a conservative cellular process that shows link with many other AD-related pathways and is crucial for maintenance of the correct cellular balance by degrading AD-associated pathogenic proteins. Considering the central role of autophagy in AD and its interplay with many other pathways, the finest therapeutic strategy to fight against AD is the use of autophagy as a target. As an essential step in this direction, this comprehensive review represents recent findings on the individual AD-related signaling pathways, describes key features of these pathways and their cross-talk with autophagy, represents current drug development, and introduces some of the multitarget beneficial approaches and strategies for the therapeutic intervention of AD.
Collapse
Affiliation(s)
- Kundlik Gadhave
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Deepak Kumar
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Vladimir N. Uversky
- Department of Molecular Medicine and Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| |
Collapse
|
41
|
Zhumakayeva S, Muravlyova L, Sirota V, Molotov-Luchansky V, Bakirova R, Kabildina N, Mkhitaryan X, Zhumakayeva A. The Reactive Carbonyl Derivatives of Proteins, Methylglyoxal, and Malondialdehyde in Blood of Women with Breast Cancer. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.5564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Every year 1.5 million women in the world are diagnosed with breast cancer (BC). In 2018, more than 260,000 new cases of cancer and more than 40,000 deaths due to this disease were detected. At the same time, in Kazakhstan, an intensive indicator of the incidences of BC in 2018 amounted to 25.3% per population of 100 thousand people (2017–24.5%) with a growth rate of 3.1%, which in absolute numbers are 4,648 new cases per year. In terms of mortality, BC ranks third after lung and stomach cancer (6.8%).
AIM: This necessitates a detailed study of the molecular mechanisms that underlie the development and progression of BC. One of the mechanisms of carcinogenesis is oxidative stress (OS). An increase in malondialdehyde (MDA) levels was detected in the early stages of cancer. It was suggested that MDA, due to its high cytotoxicity, acts as a promoter of the tumor and cocarcinogen agent.
METHODS: Therefore, violation of the parameters of OS in BC is in no doubt. However, according to the literature data analysis, these results are ambiguous and contradictory. There are no studies on a comprehensive assessment of the oxidative destruction of lipids, proteins, and nucleic acids in BC.
CONCLUSION: The nature and direction of changes in various components of OS in patients with BC have not been adequately studied, which is necessary for a correct assessment of the involvement of OS in the mechanism of the pathological process and determination of a sensitive marker of the risk of BC or its progression.
Collapse
|
42
|
Maksoud S. The Role of the Ubiquitin Proteasome System in Glioma: Analysis Emphasizing the Main Molecular Players and Therapeutic Strategies Identified in Glioblastoma Multiforme. Mol Neurobiol 2021; 58:3252-3269. [PMID: 33665742 PMCID: PMC8260465 DOI: 10.1007/s12035-021-02339-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022]
Abstract
Gliomas constitute the most frequent tumors of the brain. High-grade gliomas are characterized by a poor prognosis caused by a set of attributes making treatment difficult, such as heterogeneity and cell infiltration. Additionally, there is a subgroup of glioma cells with properties similar to those of stem cells responsible for tumor recurrence after treatment. Since proteasomal degradation regulates multiple cellular processes, any mutation causing disturbances in the function or expression of its elements can lead to various disorders such as cancer. Several studies have focused on protein degradation modulation as a mechanism of glioma control. The ubiquitin proteasome system is the main mechanism of cellular proteolysis that regulates different events, intervening in pathological processes with exacerbating or suppressive effects on diseases. This review analyzes the role of proteasomal degradation in gliomas, emphasizing the elements of this system that modulate different cellular mechanisms in tumors and discussing the potential of distinct compounds controlling brain tumorigenesis through the proteasomal pathway.
Collapse
Affiliation(s)
- Semer Maksoud
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
43
|
An X, Wei Z, Ran B, Tian H, Gu H, Liu Y, Cui H, Zhu S. Histone Deacetylase Inhibitor Trichostatin A Suppresses Cell Proliferation and Induces Apoptosis by Regulating the PI3K/AKT Signalling Pathway in Gastric Cancer Cells. Anticancer Agents Med Chem 2021; 20:2114-2124. [PMID: 32593284 DOI: 10.2174/1871520620666200627204857] [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: 12/13/2019] [Revised: 03/28/2020] [Accepted: 04/13/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Gastric cancer, a common malignant tumour worldwide, has a relatively poor prognosis and is a serious threat to human health. Histone Deacetylase Inhibitors (HDACi) are anticancer agents that are known to affect the cell growth of different cancer types. Trichostatin A (TSA) selectively inhibits the class I and II mammalian Histone Deacetylase (HDAC) family enzymes and regulates many cell processes. Still, the underlying mechanisms of HDACs are not fully understood in gastric cancer. OBJECTIVE This study aims to investigate the antitumor effect and the mechanism of growth modulation of gastric cancer cells by TSA. METHODS The cell proliferation of gastric cancer cells was measured by MTT and BrdU immunofluorescence assays. Soft agar assay was used to detect the colony formation ability of gastric cancer cells. Flow cytometry was used to examine cell cycle and apoptosis. Western blot was employed to detect protein expression of target factors. RESULTS TSA inhibits the proliferation of MKN-45 and SGC-7901 cells and leads to significant repression of colony number and size. Flow cytometry assays show TSA induces cell cycle arrest at G1 phase and apoptosis, and TSA effects the expression of related factors in the mitochondrial apoptotic signalling and cell cycle-related regulatory pathways. Furthermore, TSA increased histone H3K27 acetylation and downregulated the expression of PI3K and p-AKT. CONCLUSION Downregulating PI3K/AKT pathway activation is involved in TSA-mediated proliferation inhibition of gastric cancer.
Collapse
Affiliation(s)
- Xinli An
- School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Zekun Wei
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine,
Chongqing 400716, China
| | - Botian Ran
- School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Hao Tian
- School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Hongyu Gu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine,
Chongqing 400716, China
| | - Yan Liu
- School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine,
Chongqing 400716, China
| | - Shunqin Zhu
- School of Life Sciences, Southwest University, Chongqing, 400715, China,State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, China,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine,
Chongqing 400716, China
| |
Collapse
|
44
|
Samart P, Luanpitpong S, Rojanasakul Y, Issaragrisil S. O-GlcNAcylation homeostasis controlled by calcium influx channels regulates multiple myeloma dissemination. J Exp Clin Cancer Res 2021; 40:100. [PMID: 33726758 PMCID: PMC7968185 DOI: 10.1186/s13046-021-01876-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 02/11/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Multiple myeloma (MM) cell motility is a critical step during MM dissemination throughout the body, but how it is regulated remains largely unknown. As hypercalcemia is an important clinical feature of MM, high calcium (Ca2+) and altered Ca2+ signaling could be a key contributing factor to the pathological process. METHODS Bioinformatics analyses were employed to assess the clinical significance of Ca2+ influx channels in clinical specimens of smoldering and symptomatic MM. Functional and regulatory roles of influx channels and downstream signaling in MM cell migration and invasion were conducted and experimental MM dissemination was examined in a xenograft mouse model using in vivo live imaging and engraftment analysis. RESULTS Inhibition of TRPM7, ORAI1, and STIM1 influx channels, which are highly expressed in MM patients, and subsequent blockage of Ca2+ influx by CRISPR/Cas9 and small molecule inhibitors, effectively inhibit MM cell migration and invasion, and attenuate the experimental MM dissemination. Mechanistic studies reveal a nutrient sensor O-GlcNAcylation as a downstream regulator of Ca2+ influx that specifically targets cell adhesion molecules. Hyper-O-GlcNAcylation following the inhibition of Ca2+ influx channels induces integrin α4 and integrin β7 downregulation via ubiquitin-proteasomal degradation and represses the aggressive MM phenotype. CONCLUSIONS Our findings unveil a novel regulatory mechanism of MM cell motility via Ca2+ influx/O-GlcNAcylation axis that directly targets integrin α4 and integrin β7, providing mechanistic insights into the pathogenesis and progression of MM and demonstrating potential predictive biomarkers and therapeutic targets for advanced MM.
Collapse
Affiliation(s)
- Parinya Samart
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Siriraj Hospital, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Yon Rojanasakul
- WVU Cancer Institute and Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Siriraj Hospital, Bangkoknoi, Bangkok, 10700, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Bangkok Hematology Center, Wattanosoth Hospital, BDMS Center of Excellence for Cancer, Bangkok, Thailand
| |
Collapse
|
45
|
Mishra N, Mishra S. Exendin-4 - A potential therapeutic for type 2 diabetes-linked cervical cancer? EBioMedicine 2021; 65:103273. [PMID: 33714887 PMCID: PMC7966980 DOI: 10.1016/j.ebiom.2021.103273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 11/25/2022] Open
Affiliation(s)
- Nivida Mishra
- Faculty of Engineering, University of Manitoba, Manitoba, Winnipeg, Canada
| | - Suresh Mishra
- Department of Internal Medicine, Faculty of Health Sciences, University of Manitoba, Manitoba, Winnipeg, Canada.
| |
Collapse
|
46
|
Erokhov PA, Kulikov AM, Karpova YD, Rodoman GV, Sumedi IR, Goncharov AL, Razbirin DV, Gorelova VS, Sharova NP, Astakhova TM. Proteasomes in Patient Rectal Cancer and Different Intestine Locations: Where Does Proteasome Pool Change? Cancers (Basel) 2021; 13:1108. [PMID: 33807574 PMCID: PMC7961961 DOI: 10.3390/cancers13051108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/01/2021] [Indexed: 01/12/2023] Open
Abstract
A special problem in the surgery of rectal cancer is connected with a need for appropriate removal of intestine parts, along with the tumor, including the fragment close to the sphincter. To determine the length of fragments to remove, it is necessary to reveal areas without changes in molecule functioning, specific for tumor. The purpose of the present study was to investigate functioning the proteasomes, the main actors in protein hydrolysis, in patient rectal adenocarcinoma and different intestine locations. Chymotrypsin-like and caspase-like activities, open to complex influence of different factors, were analyzed in 43-54 samples by Suc-LLVY-AMC- and Z-LLE-AMC-hydrolysis correspondingly. Both activities may be arranged by the decrease in the location row: cancer→adjacent tissue→proximal (8-20 cm from tumor) and distal (2 and 4 cm from tumor) sides. These activities did not differ noticeably in proximal and distal locations. Similar patterns were detected for the activities and expression of immune subunits LMP2 and LMP7 and expression of 19S and PA28αβ activators. The largest changes in tumor were related to proteasome subtype containing LMP2 and PA28αβ that was demonstrated by native electrophoresis. Thus, the results indicate a significance of subtype LMP2-PA28αβ for tumor and absence of changes in proteasome pool in distal fragments of 2-4 cm from tumor.
Collapse
Affiliation(s)
- Pavel A. Erokhov
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (P.A.E.); (A.M.K.); (Y.D.K.); (V.S.G.); (T.M.A.)
| | - Alexey M. Kulikov
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (P.A.E.); (A.M.K.); (Y.D.K.); (V.S.G.); (T.M.A.)
| | - Yaroslava D. Karpova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (P.A.E.); (A.M.K.); (Y.D.K.); (V.S.G.); (T.M.A.)
| | - Grigory V. Rodoman
- Pirogov Russian National Research Medical University of Ministry of Health of Russian Federation, 1 Ostrovityanov Street, 117997 Moscow, Russia; (G.V.R.); (I.R.S.); (A.L.G.); (D.V.R.)
| | - Ilia R. Sumedi
- Pirogov Russian National Research Medical University of Ministry of Health of Russian Federation, 1 Ostrovityanov Street, 117997 Moscow, Russia; (G.V.R.); (I.R.S.); (A.L.G.); (D.V.R.)
| | - Artem L. Goncharov
- Pirogov Russian National Research Medical University of Ministry of Health of Russian Federation, 1 Ostrovityanov Street, 117997 Moscow, Russia; (G.V.R.); (I.R.S.); (A.L.G.); (D.V.R.)
| | - Dmitry V. Razbirin
- Pirogov Russian National Research Medical University of Ministry of Health of Russian Federation, 1 Ostrovityanov Street, 117997 Moscow, Russia; (G.V.R.); (I.R.S.); (A.L.G.); (D.V.R.)
| | - Vera S. Gorelova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (P.A.E.); (A.M.K.); (Y.D.K.); (V.S.G.); (T.M.A.)
| | - Natalia P. Sharova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (P.A.E.); (A.M.K.); (Y.D.K.); (V.S.G.); (T.M.A.)
| | - Tatiana M. Astakhova
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (P.A.E.); (A.M.K.); (Y.D.K.); (V.S.G.); (T.M.A.)
| |
Collapse
|
47
|
Mao D, Cao H, Shi M, Wang CC, Kwong J, Li JJX, Hou Y, Ming X, Lee HM, Tian XY, Wong CK, Chow E, Kong APS, Lui VWY, Chan PKS, Chan JCN. Increased co-expression of PSMA2 and GLP-1 receptor in cervical cancer models in type 2 diabetes attenuated by Exendin-4: A translational case-control study. EBioMedicine 2021; 65:103242. [PMID: 33684886 PMCID: PMC7938253 DOI: 10.1016/j.ebiom.2021.103242] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) increases the risk of many types of cancer. Dysregulation of proteasome-related protein degradation leads to tumorigenesis, while Exendin-4, a glucagon-like peptide 1 receptor (GLP-1R) agonist, possesses anti-cancer effects. METHODS We explored the co-expression of proteasome alpha 2 subunit (PSMA2) and GLP-1R in the Cancer Genome Atlas (TCGA) database and human cervical cancer specimens, supplemented by in vivo and in vitro studies using multiple cervical cancer cell lines. FINDINGS PSMA2 expression was increased in 12 cancer types in TCGA database and cervical cancer specimens from patients with T2D (T2D vs non-T2D: 3.22 (95% confidence interval CI: 1.38, 5.05) vs 1.00 (0.66, 1.34) fold change, P = 0.01). psma2-shRNA decreased cell proliferation in vitro, and tumour volume and Ki67 expression in vivo. Exendin-4 decreased psma2 expression, tumour volume and Ki67 expression in vivo. There was no change in GLP-1R expression in 12 cancer types in TCGA database. However, GLP-1R expression (T2D vs non-T2D: 5.49 (3.0, 8.1) vs 1.00 (0.5, 1.5) fold change, P < 0.001) was increased and positively correlated with PSMA2 expression in T2D-related (r = 0.68) but not in non-T2D-related cervical cancer specimens. This correlation was corroborated by in vitro experiments where silencing glp-1r decreased psma2 expression. Exendin-4 attenuated phospho-p65 and -IκB expression in the NF-κB pathway. INTERPRETATION PSMA2 and GLP-1R expression in T2D-related cervical cancer specimens was increased and positively correlated, suggesting hyperglycaemia might promote cancer growth by increasing PSMA2 expression which could be attenuated by Exendin-4. FUNDING This project was supported by Postdoctoral Fellowship Scheme, Direct Grant, Diabetes Research and Education Fund from the Chinese University of Hong Kong (CUHK).
Collapse
Affiliation(s)
- Dandan Mao
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Huanyi Cao
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Mai Shi
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Joseph Kwong
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Joshua Jing Xi Li
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Yong Hou
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Xing Ming
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Heung Man Lee
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chun Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Alice Pik Shan Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Vivian Wai Yan Lui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Paul Kay Sheung Chan
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Juliana Chung Ngor Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China.
| |
Collapse
|
48
|
Liao Y, Lupiani B, Izumiya Y, Reddy SM. Marek's disease virus Meq oncoprotein interacts with chicken HDAC 1 and 2 and mediates their degradation via proteasome dependent pathway. Sci Rep 2021; 11:637. [PMID: 33437016 PMCID: PMC7803728 DOI: 10.1038/s41598-020-80792-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/28/2020] [Indexed: 12/02/2022] Open
Abstract
Marek's disease virus (MDV) encodes a basic-leucine zipper (BZIP) protein, Meq, which is considered the major MDV oncoprotein. It has been reported that the oncogenicity of Meq is associated with its interaction with C-terminal binding protein 1 (CtBP), which is also an interaction partner of Epstein-Barr virus encoded EBNA3A and EBNA3C oncoproteins. Since both EBNA3C and CtBP interact with histone deacetylase 1 (HDAC1) and HDAC2, we examined whether Meq shares this interaction with chicken HDAC1 (chHDAC1) and chHDAC2. Using confocal microscopy analysis, we show that Meq co-localizes with chHDAC1 and chHDAC2 in the nuclei of MDV lymphoblastoid tumor cells. In addition, immunoprecipitation assays demonstrate that Meq interacts with chHDAC1 and chHDAC2 in transfected cells and MDV lymphoblastoid tumor cells. Using deletion mutants, interaction domains were mapped to the N-terminal dimerization domain of chHDAC1 and chHDAC2, and the BZIP domain of Meq. Our results further demonstrate that this interaction mediates the degradation of chHDAC1 and chHDAC2 via the proteasome dependent pathway. In addition, our results show that Meq also induces the reduction of global ubiquitinated proteins through a proteasome dependent pathway. In conclusion, our results provide evidence that Meq interacts with chHDAC1 and chHDAC2, and induces their proteasome dependent degradation.
Collapse
Affiliation(s)
- Yifei Liao
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, MS4467, TAMU, College Station, TX, 77843, USA
| | - Blanca Lupiani
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, MS4467, TAMU, College Station, TX, 77843, USA
| | - Yoshihiro Izumiya
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Sanjay M Reddy
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, MS4467, TAMU, College Station, TX, 77843, USA.
| |
Collapse
|
49
|
Schukur L, Zimmermann T, Niewoehner O, Kerr G, Gleim S, Bauer-Probst B, Knapp B, Galli GG, Liang X, Mendiola A, Reece-Hoyes J, Rapti M, Barbosa I, Reschke M, Radimerski T, Thoma CR. Identification of the HECT E3 ligase UBR5 as a regulator of MYC degradation using a CRISPR/Cas9 screen. Sci Rep 2020; 10:20044. [PMID: 33208877 PMCID: PMC7676242 DOI: 10.1038/s41598-020-76960-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
MYC oncoprotein is a multifunctional transcription factor that regulates the expression of a large number of genes involved in cellular growth, proliferation and metabolism. Altered MYC protein level lead to cellular transformation and tumorigenesis. MYC is deregulated in > 50% of human cancers, rendering it an attractive drug target. However, direct inhibition of this class of proteins using conventional small molecules is challenging due to their intrinsically disordered state. To discover novel posttranslational regulators of MYC protein stability and turnover, we established a genetic screen in mammalian cells by combining a fluorescent protein-based MYC abundance sensor, CRISPR/Cas9-based gene knockouts and next-generation sequencing. Our screen identifies UBR5, an E3 ligase of the HECT-type family, as a novel regulator of MYC degradation. Even in the presence of the well-described and functional MYC ligase, FBXW7, UBR5 depletion leads to accumulation of MYC in cells. We demonstrate interaction of UBR5 with MYC and reduced K48-linked ubiquitination of MYC upon loss of UBR5 in cells. Interestingly, in cancer cell lines with amplified MYC expression, depletion of UBR5 resulted in reduced cell survival, as a consequence of MYC stabilization. Finally, we show that MYC and UBR5 are co-amplified in more than 40% of cancer cells and that MYC copy number amplification correlates with enhanced transcriptional output of UBR5. This suggests that UBR5 acts as a buffer in MYC amplified settings and protects these cells from apoptosis.
Collapse
Affiliation(s)
- Lina Schukur
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland. .,Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Tamara Zimmermann
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Ole Niewoehner
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA
| | - Grainne Kerr
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Scott Gleim
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA
| | - Beatrice Bauer-Probst
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Britta Knapp
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA
| | - Giorgio G Galli
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Xiaoyou Liang
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA
| | - Angelica Mendiola
- Genomics Institute of the Novartis Research Foundation, LaJolla, USA
| | - John Reece-Hoyes
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA
| | - Melivoia Rapti
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Ines Barbosa
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Markus Reschke
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland
| | - Thomas Radimerski
- Novartis Institutes for Biomedical Research (NIBR) Oncology, Novartis, Basel, Switzerland.,Basilea Pharmaceutica, Basel, Switzerland
| | - Claudio R Thoma
- NIBR Chemical Biology and Therapeutics, Novartis, Cambridge, USA.
| |
Collapse
|
50
|
Tundo GR, Sbardella D, Santoro AM, Coletta A, Oddone F, Grasso G, Milardi D, Lacal PM, Marini S, Purrello R, Graziani G, Coletta M. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges. Pharmacol Ther 2020; 213:107579. [PMID: 32442437 PMCID: PMC7236745 DOI: 10.1016/j.pharmthera.2020.107579] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023]
Abstract
Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | | | - A M Santoro
- CNR, Institute of Crystallography, Catania, Italy
| | - A Coletta
- Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - F Oddone
- IRCCS-Fondazione Bietti, Rome, Italy
| | - G Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - D Milardi
- CNR, Institute of Crystallography, Catania, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - R Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|