1
|
Van der Vreken A, Thery F, Tu C, Mwangi K, Meulewaeter S, De Beck L, Janssens E, De Veirman K, Vanderkerken K, De Bruyne E, Franceschini L, Impens F, Verbeke R, Lentacker I, Menu E, Breckpot K. Immunopeptidomics identified antigens for mRNA-lipid nanoparticle vaccines with alpha-galactosylceramide in multiple myeloma therapy. J Immunother Cancer 2025; 13:e010673. [PMID: 40300855 PMCID: PMC12049997 DOI: 10.1136/jitc-2024-010673] [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: 09/27/2024] [Accepted: 04/05/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Invariant natural killer T (iNKT) cells and CD8+ T cells are key in the immune response against multiple myeloma (MM), a largely incurable blood cancer. Immunization is a promising strategy to activate these T cell populations. To our knowledge, immunization with messenger RNA (mRNA) and the iNKT agonist, α-galactosylceramide (αGC), has not been studied in MM, as knowledge on clinically relevant antigens in preclinical MM models is lacking. METHODS Microarray data and immunopeptidomics (imPep) were used to identify candidate antigens for immunization in 5TMM models. Galsomes, lipid nanoparticles containing antigen mRNA and αGC were used to immunize 5T33MM-bearing mice. This treatment was combined with a CD40 agonist. Tumor burden and activation of iNKT cells and CD8+ T cells were studied using M-protein electrophoresis, flow cytometry and ELISA. RESULTS RNA transcripts revealed survivin as a candidate antigen. Prime-boost Galsomes therapy targeting survivin significantly reduced M-protein levels despite low survivin-specific T cell responses. Further analysis showed potential T cell fratricide. ImPep revealed HSP60, Idiotype, PICALM and EF1A1 as candidate antigens. Prime-boost therapy with Galsomes targeting these antigens reduced MM growth significantly when combined with a CD40 agonist, coinciding with significantly improved antigen presentation, costimulation and cytotoxicity of iNKT cells and CD8+ T cells. CONCLUSION These findings highlight the potential of Galsomes, an mRNA vaccine designed to activate CD8+ T cells and iNKT cells, for MM therapy, and emphasize the importance of combinatorial approaches, addressing immune anergy for effective MM immunotherapies.
Collapse
Affiliation(s)
- Arne Van der Vreken
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fabien Thery
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Chenggong Tu
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kevin Mwangi
- Department of Pharmaceutics, Laboratory of Physical Pharmacy and General Biochemistry, Ghent Research Group on Nanomedicines, Universiteit Gent, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Universiteit Gent, Ghent, Belgium
| | - Sofie Meulewaeter
- Department of Pharmaceutics, Laboratory of Physical Pharmacy and General Biochemistry, Ghent Research Group on Nanomedicines, Universiteit Gent, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Universiteit Gent, Ghent, Belgium
| | - Lien De Beck
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Edith Janssens
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kim De Veirman
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karin Vanderkerken
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lorenzo Franceschini
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Rein Verbeke
- Department of Pharmaceutics, Laboratory of Physical Pharmacy and General Biochemistry, Ghent Research Group on Nanomedicines, Universiteit Gent, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Universiteit Gent, Ghent, Belgium
| | - Ine Lentacker
- Department of Pharmaceutics, Laboratory of Physical Pharmacy and General Biochemistry, Ghent Research Group on Nanomedicines, Universiteit Gent, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Universiteit Gent, Ghent, Belgium
| | - Eline Menu
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
2
|
Munguia-Galaviz FJ, Gutierrez-Mercado YK, Miranda-Diaz AG, Portilla de Buen E, Flores-Soto ME, Echavarria R. Cardiac transcriptomic changes induced by early CKD in mice reveal novel pathways involved in the pathogenesis of Cardiorenal syndrome type 4. Heliyon 2024; 10:e27468. [PMID: 38509984 PMCID: PMC10950824 DOI: 10.1016/j.heliyon.2024.e27468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/26/2023] [Accepted: 02/29/2024] [Indexed: 03/22/2024] Open
Abstract
Background Cardiorenal syndrome (CRS) type 4 is prevalent among the chronic kidney disease (CKD) population, with many patients dying from cardiovascular complications. However, limited data regarding cardiac transcriptional changes induced early by CKD is available. Methods We used a murine unilateral ureteral obstruction (UUO) model to evaluate renal damage, cardiac remodeling, and transcriptional regulation at 21 days post-surgery through histological analysis, RT-qPCR, RNA-seq, and bioinformatics. Results UUO leads to significant kidney injury, low uremia, and pathological cardiac remodeling, evidenced by increased collagen deposition and smooth muscle alpha-actin 2 expression. RNA-seq analysis identified 76 differentially expressed genes (DEGs) in UUO hearts. Upregulated DEGs were significantly enriched in cell cycle and cell division pathways, immune responses, cardiac repair, inflammation, proliferation, oxidative stress, and apoptosis. Gene Set Enrichment Analysis further revealed mitochondrial oxidative bioenergetic pathways, autophagy, and peroxisomal pathways are downregulated in UUO hearts. Vimentin was also identified as an UUO-upregulated transcript. Conclusions Our results emphasize the relevance of extensive transcriptional changes, mitochondrial dysfunction, homeostasis deregulation, fatty-acid metabolism alterations, and vimentin upregulation in CRS type 4 development.
Collapse
Affiliation(s)
- Francisco Javier Munguia-Galaviz
- Departamento de Fisiologia, CUCS, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Division de Ciencias de la Salud, CUSUR, Universidad de Guadalajara, Ciudad Guzman 49000, Jalisco, Mexico
| | | | | | - Eliseo Portilla de Buen
- Division de Investigacion Quirurgica, Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico
| | - Mario Eduardo Flores-Soto
- Division de Neurociencias, Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico
| | - Raquel Echavarria
- CONAHCYT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
3
|
Orozco RC, Marquardt K, Pratumchai I, Shaikh AF, Mowen K, Domissy A, Teijaro JR, Sherman LA. Autoimmunity-associated allele of tyrosine phosphatase gene PTPN22 enhances anti-viral immunity. PLoS Pathog 2024; 20:e1012095. [PMID: 38512979 PMCID: PMC10987006 DOI: 10.1371/journal.ppat.1012095] [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: 04/26/2023] [Revised: 04/02/2024] [Accepted: 03/04/2024] [Indexed: 03/23/2024] Open
Abstract
The 1858C>T allele of the tyrosine phosphatase PTPN22 is present in 5-10% of the North American population and is strongly associated with numerous autoimmune diseases. Although research has been done to define how this allele potentiates autoimmunity, the influence PTPN22 and its pro-autoimmune allele has in anti-viral immunity remains poorly defined. Here, we use single cell RNA-sequencing and functional studies to interrogate the impact of this pro-autoimmune allele on anti-viral immunity during Lymphocytic Choriomeningitis Virus clone 13 (LCMV-cl13) infection. Mice homozygous for this allele (PEP-619WW) clear the LCMV-cl13 virus whereas wildtype (PEP-WT) mice cannot. This is associated with enhanced anti-viral CD4 T cell responses and a more immunostimulatory CD8α- cDC phenotype. Adoptive transfer studies demonstrated that PEP-619WW enhanced anti-viral CD4 T cell function through virus-specific CD4 T cell intrinsic and extrinsic mechanisms. Taken together, our data show that the pro-autoimmune allele of Ptpn22 drives a beneficial anti-viral immune response thereby preventing what is normally a chronic virus infection.
Collapse
Affiliation(s)
- Robin C. Orozco
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - Kristi Marquardt
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Isaraphorn Pratumchai
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Anam Fatima Shaikh
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - Kerri Mowen
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Alain Domissy
- Genomics Core, Scripps Research, La Jolla, California, United States of America
| | - John R. Teijaro
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Linda A. Sherman
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, United States of America
| |
Collapse
|
4
|
Zhang Q, Chen X, Hu Y, Zhou T, Du M, Xu R, Chen Y, Tang P, Chen Z, Li J. BIRC5 Inhibition Is Associated with Pyroptotic Cell Death via Caspase3-GSDME Pathway in Lung Adenocarcinoma Cells. Int J Mol Sci 2023; 24:14663. [PMID: 37834111 PMCID: PMC10572361 DOI: 10.3390/ijms241914663] [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: 07/27/2023] [Revised: 08/26/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is a prevalent type of thoracic cancer with a poor prognosis and high mortality rate. However, the exact pathogenesis of this cancer is still not fully understood. One potential factor that can contribute to the development of lung adenocarcinoma is DNA methylation, which can cause changes in chromosome structure and potentially lead to the formation of tumors. The baculoviral IAP repeat containing the 5 (BIRC5) gene encodes the Survivin protein, which is a multifunctional gene involved in cell proliferation, migration, and invasion of tumor cells. This gene is elevated in various solid tumors, but its specific role and mechanism in lung adenocarcinoma are not well-known. To identify the potential biomarkers associated with lung adenocarcinoma, we screened the methylation-regulated differentially expressed genes (MeDEGs) of LUAD via bioinformatics analysis. Gene ontology (GO) process and the Kyoto Encyclopedia of Genes and Genomes (KEGG) were applied to investigate the biological function and pathway of MeDEGs. A protein-protein interaction (PPI) network was employed to explore the key module and screen hub genes. We screened out eight hub genes whose products are aberrantly expressed, and whose DNA methylation modification level is significantly changed in lung adenocarcinoma. BIRC5 is a bona fide marker which was remarkably up-regulated in tumor tissues. Flow cytometry analysis, lactate dehydrogenase release (LDH) assay and Micro-PET imaging were performed in A549 cells and a mouse xenograft tumor to explore the function of BIRC5 in cell death of lung adenocarcinoma. We found that BIRC5 was up-regulated and related to a high mortality rate in lung adenocarcinoma patients. Mechanically, the knockdown of BIRC5 inhibited the proliferation of A549 cells and induced pyroptosis via caspase3/GSDME signaling. Our findings have unraveled that BIRC5 holds promise as a novel biomarker and therapeutic target for lung adenocarcinoma. Additionally, we have discovered a novel pathway in which BIRC5 inhibition can induce pyroptosis through the caspase3-GSDME pathway in lung adenocarcinoma cells.
Collapse
Affiliation(s)
- Qingwei Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin 150081, China
| | - Ximing Chen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yingying Hu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Tong Zhou
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Menghan Du
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Run Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yongchao Chen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Pingping Tang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Zhouxiu Chen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Jiamin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
5
|
Loredan DG, Devlin JC, Lacey KA, Howard N, Chen Z, Zwack EE, Lin JD, Ruggles KV, Khanna KM, Torres VJ, Loke P. Single-Cell Analysis of CX3CR1+ Cells Reveals a Pathogenic Role for BIRC5+ Myeloid Proliferating Cells Driven by Staphylococcus aureus Leukotoxins. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:836-843. [PMID: 37466391 PMCID: PMC10450158 DOI: 10.4049/jimmunol.2300166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/30/2023] [Indexed: 07/20/2023]
Abstract
Our previous studies identified a population of stem cell-like proliferating myeloid cells within inflamed tissues that could serve as a reservoir for tissue macrophages to adopt different activation states depending on the microenvironment. By lineage-tracing cells derived from CX3CR1+ precursors in mice during infection and profiling by single-cell RNA sequencing, in this study, we identify a cluster of BIRC5+ myeloid cells that expanded in the liver during chronic infection with either the parasite Schistosoma mansoni or the bacterial pathogen Staphylococcus aureus. In the absence of tissue-damaging toxins, S. aureus infection does not elicit these BIRC5+ cells. Moreover, deletion of BIRC5 from CX3CR1-expressing cells results in improved survival during S. aureus infection. Hence the combination of single-cell RNA sequencing and genetic fate-mapping CX3CR1+ cells revealed a toxin-dependent pathogenic role for BIRC5 in myeloid cells during S. aureus infection.
Collapse
Affiliation(s)
- Denis G. Loredan
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY
| | - Joseph C. Devlin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Keenan A. Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Nina Howard
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Ze Chen
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Erin E. Zwack
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Jian-Da Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei City, Taiwan
- Center for Computational and Systems Biology, National Taiwan University, Taipei City, Taiwan
| | - Kelly V. Ruggles
- Institute of Systems Genetics, New York University Grossman School of Medicine, New York, NY
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Kamal M. Khanna
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY
| | - P’ng Loke
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
6
|
Loredan DG, Devlin JC, Lacey KA, Howard N, Chen Z, Zwack EE, Lin JD, Ruggles KV, Khanna KM, Torres VJ, Loke PN. Single-cell analysis of CX3CR1 + cells reveal a pathogenic role for BIRC5 + myeloid proliferating cells driven by Staphylococcus aureus leukotoxins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.529760. [PMID: 36909517 PMCID: PMC10002671 DOI: 10.1101/2023.02.27.529760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Our previous studies identified a population of stem cell-like proliferating myeloid cells within inflamed tissues that could serve as a reservoir for tissue macrophages to adopt different activation states depending on the microenvironment. By lineage tracing cells derived from CX3CR1 + precursors in mice during infection and profiling by scRNA-seq, here we identify a cluster of BIRC5 + myeloid cells that expanded in the liver during either chronic infection with the parasite Schistosoma mansoni or the bacterial pathogen Staphylococcus aureus . In the absence of tissue damaging toxins, S. aureus infection does not elicit these BIRC5 + cells. Moreover, deletion of BIRC5 from CX3CR1 expressing cells results in improved survival during S. aureus infection. Hence, the combination of scRNA-Seq and genetic fate mapping CX3CR1 + cells revealed a toxin dependent pathogenic role for BIRC5 in myeloid cells during S. aureus infection.
Collapse
|
7
|
Chaithongyot S, Naumann M. Helicobacter pylori-induced reactive oxygen species direct turnover of CSN-associated STAMBPL1 and augment apoptotic cell death. Cell Mol Life Sci 2022; 79:86. [PMID: 35066747 PMCID: PMC8784504 DOI: 10.1007/s00018-022-04135-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/11/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023]
Abstract
Deubiquitinylases (DUBs) are central regulators of the ubiquitin system involved in protein regulation and cell signalling and are important for a variety of physiological processes. Most DUBs are cysteine proteases, and few other proteases are metalloproteases of the JAB1/MPN +/MOV34 protease family (JAMM). STAM-binding protein like 1 (STAMBPL1), a member of the JAMM family, cleaves ubiquitin bonds and has a function in regulating cell survival, Tax-mediated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation and epithelial-mesenchymal transition. However, the molecular mechanism by which STAMBPL1 influences cell survival is not well defined, especially with regard to its deubiquitinylation function. Here, we show that reactive oxygen species (ROS) induced by chemotherapeutic agents or the human microbial pathogen Helicobacter pylori can induce cullin 1-RING ubiquitin ligase (CRL1) and 26S proteasome-dependent degradation STAMBPL1. Interestingly, STAMBPL1 has a direct interaction with the constitutive photomorphogenic 9 (COP9 or CSN) signalosome subunits CSN5 and CSN6. The interaction with the CSN is required for the stabilisation and function of the STAMBPL1 protein. In addition, STAMBPL1 deubiquitinylates the anti-apoptotic protein Survivin and thus ameliorates cell survival. In summary, our data reveal a previously unknown mechanism by which the deubiquitinylase STAMBPL1 and the E3 ligase CRL1 balance the level of Survivin degradation and thereby determine apoptotic cell death. In response to genotoxic stress, the degradation of STAMBPL1 augments apoptotic cell death. This new mechanism may be useful to develop therapeutic strategies targeting STAMBPL1 in tumours that have high STAMBPL1 and Survivin protein levels.
Collapse
|