1
|
Li YY, Meng FL, Zhou SY, Du JM, Li WJ, Liu QY, Wu L, Zhao MM, Jin Y, Zhang QY, Li Y, Su GH. CDX1 improves nicotine induced cardiac fibroblasts activation and cardiomyocyte hypertrophy by alleviating autophagic flux impairment through modulation of LAPTM4B. Sci Rep 2025; 15:9985. [PMID: 40121311 PMCID: PMC11929911 DOI: 10.1038/s41598-025-94160-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Nicotine-induced impairment of autophagic flux promotes the onset of myocardial remodelling, thereby exacerbating heart failure. In this study, we investigated the role and molecular mechanisms of the transcription factor CDX1 in cardiac fibroblasts (CFs) activation and cardiomyocyte hypertrophy induced by nicotine. We found that CDX1 expression was increased in response to nicotine. However, a decrease in CDX1 further exacerbated the nicotine-induced blockade of autophagic flux, thereby aggravating CFs activation and cardiomyocyte hypertrophy. This effect was attributed to the suppression of the autophagic regulator LAPTM4B transcription by CDX1 and the subsequent activation of the mTOR pathway. In contrast, CDX1 overexpression promoted LAPTM4B expression, resulting in the opposite effect. In conclusion, our study demonstrated that CDX1/LAPTM4B axis could alleviate nicotine-induced autophagy flux impairment by inhibiting mTORC1 pathway activation, thereby alleviating CFs activation and cardiomyocyte hypertrophy, and exerting cardioprotective functions.
Collapse
Affiliation(s)
- Yue-Yan Li
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China
- Research Center for Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fan-Liang Meng
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Si-Yuan Zhou
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Jia-Min Du
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Wen-Jing Li
- Research Center for Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qi-Yun Liu
- Department of Cardiology, Shandong Second Medical University, Weifang, China
| | - Lei Wu
- Research Center for Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Meng-Meng Zhao
- Research Center for Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yi Jin
- Research Center for Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qun-Ye Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Ying Li
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China.
- Research Center for Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Guo-Hai Su
- Department of Cardiology, Jinan Central Hospital, Shandong University, Jinan, China.
- Research Center for Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
2
|
Brombacher EC, Patente TA, van der Ham AJ, Moll TJ, Otto F, Verheijen FW, Zaal EA, de Ru AH, Tjokrodirijo RT, Berkers CR, van Veelen PA, Guigas B, Everts B. AMPK activation induces RALDH+ tolerogenic dendritic cells by rewiring glucose and lipid metabolism. J Cell Biol 2024; 223:e202401024. [PMID: 39115541 PMCID: PMC11310580 DOI: 10.1083/jcb.202401024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/30/2024] [Accepted: 07/10/2024] [Indexed: 09/13/2024] Open
Abstract
Dendritic cell (DC) activation and function are underpinned by profound changes in cellular metabolism. Several studies indicate that the ability of DCs to promote tolerance is dependent on catabolic metabolism. Yet the contribution of AMP-activated kinase (AMPK), a central energy sensor promoting catabolism, to DC tolerogenicity remains unknown. Here, we show that AMPK activation renders human monocyte-derived DCs tolerogenic as evidenced by an enhanced ability to drive differentiation of regulatory T cells, a process dependent on increased RALDH activity. This is accompanied by several metabolic changes, including increased breakdown of glycerophospholipids, enhanced mitochondrial fission-dependent fatty acid oxidation, and upregulated glucose catabolism. This metabolic rewiring is functionally important as we found interference with these metabolic processes to reduce to various degrees AMPK-induced RALDH activity as well as the tolerogenic capacity of moDCs. Altogether, our findings reveal a key role for AMPK signaling in shaping DC tolerogenicity and suggest AMPK as a target to direct DC-driven tolerogenic responses in therapeutic settings.
Collapse
Affiliation(s)
- Eline C. Brombacher
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Thiago A. Patente
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Alwin J. van der Ham
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Tijmen J.A. Moll
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Frank Otto
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Fenne W.M. Verheijen
- Department Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Esther A. Zaal
- Department Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Arnoud H. de Ru
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | | | - Celia R. Berkers
- Department Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Peter A. van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
3
|
de Kivit S, Mensink M, Kostidis S, Derks RJE, Zaal EA, Heijink M, Verleng LJ, de Vries E, Schrama E, Blomberg N, Berkers CR, Giera M, Borst J. Immune suppression by human thymus-derived effector Tregs relies on glucose/lactate-fueled fatty acid synthesis. Cell Rep 2024; 43:114681. [PMID: 39180751 DOI: 10.1016/j.celrep.2024.114681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/10/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024] Open
Abstract
Regulatory T cells (Tregs) suppress pro-inflammatory conventional T cell (Tconv) responses. As lipids impact cell signaling and function, we compare the lipid composition of CD4+ thymus-derived (t)Tregs and Tconvs. Lipidomics reveal constitutive enrichment of neutral lipids in Tconvs and phospholipids in tTregs. TNFR2-co-stimulated effector tTregs and Tconvs are both glycolytic, but only in tTregs are glycolysis and the tricarboxylic acid (TCA) cycle linked to a boost in fatty acid (FA) synthesis (FAS), supported by relevant gene expression. FA chains in tTregs are longer and more unsaturated than in Tconvs. In contrast to Tconvs, tTregs effectively use either lactate or glucose for FAS and rely on this process for proliferation. FASN and SCD1, enzymes responsible for FAS and FA desaturation, prove essential for the ability of tTregs to suppress Tconvs. These data illuminate how effector tTregs can thrive in inflamed or cancerous tissues with limiting glucose but abundant lactate levels.
Collapse
Affiliation(s)
- Sander de Kivit
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.
| | - Mark Mensink
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Sarantos Kostidis
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Rico J E Derks
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Esther A Zaal
- Division of Cell Biology, Metabolism, and Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Marieke Heijink
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Lotte J Verleng
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Evert de Vries
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Ellen Schrama
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Niek Blomberg
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Celia R Berkers
- Division of Cell Biology, Metabolism, and Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Jannie Borst
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.
| |
Collapse
|
4
|
Karakaslar EO, Severens JF, Sánchez-López E, van Veelen PA, Zlei M, van Dongen JJM, Otte AM, Halkes CJM, van Balen P, Veelken H, Reinders MJT, Griffioen M, van den Akker EB. A transcriptomic based deconvolution framework for assessing differentiation stages and drug responses of AML. NPJ Precis Oncol 2024; 8:105. [PMID: 38762545 PMCID: PMC11102519 DOI: 10.1038/s41698-024-00596-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 05/03/2024] [Indexed: 05/20/2024] Open
Abstract
The diagnostic spectrum for AML patients is increasingly based on genetic abnormalities due to their prognostic and predictive value. However, information on the AML blast phenotype regarding their maturational arrest has started to regain importance due to its predictive power for drug responses. Here, we deconvolute 1350 bulk RNA-seq samples from five independent AML cohorts on a single-cell healthy BM reference and demonstrate that the morphological differentiation stages (FAB) could be faithfully reconstituted using estimated cell compositions (ECCs). Moreover, we show that the ECCs reliably predict ex-vivo drug resistances as demonstrated for Venetoclax, a BCL-2 inhibitor, resistance specifically in AML with CD14+ monocyte phenotype. We validate these predictions using LUMC proteomics data by showing that BCL-2 protein abundance is split into two distinct clusters for NPM1-mutated AML at the extremes of CD14+ monocyte percentages, which could be crucial for the Venetoclax dosing patients. Our results suggest that Venetoclax resistance predictions can also be extended to AML without recurrent genetic abnormalities and possibly to MDS-related and secondary AML. Lastly, we show that CD14+ monocytic dominated Ven/Aza treated patients have significantly lower overall survival. Collectively, we propose a framework for allowing a joint mutation and maturation stage modeling that could be used as a blueprint for testing sensitivity for new agents across the various subtypes of AML.
Collapse
Affiliation(s)
- E Onur Karakaslar
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
- Pattern Recognition & Bioinformatics, Delft University of Technology, Delft, The Netherlands
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeppe F Severens
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
- Pattern Recognition & Bioinformatics, Delft University of Technology, Delft, The Netherlands
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Elena Sánchez-López
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, The Netherlands
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Mihaela Zlei
- Department of Flow Cytometry, Medical Laboratory, Regional Institute of Oncology, Iasi, Romania
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacques J M van Dongen
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC, USAL-CSIC-FICUS) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Annemarie M Otte
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Peter van Balen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hendrik Veelken
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marcel J T Reinders
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
- Pattern Recognition & Bioinformatics, Delft University of Technology, Delft, The Netherlands
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik B van den Akker
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands.
- Pattern Recognition & Bioinformatics, Delft University of Technology, Delft, The Netherlands.
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
5
|
Li J, Purser N, Liwocha J, Scott DC, Byers HA, Steigenberger B, Hill S, Tripathi-Giesgen I, Hinkle T, Hansen FM, Prabu JR, Radhakrishnan SK, Kirkpatrick DS, Reichermeier KM, Schulman BA, Kleiger G. Cullin-RING ligases employ geometrically optimized catalytic partners for substrate targeting. Mol Cell 2024; 84:1304-1320.e16. [PMID: 38382526 PMCID: PMC10997478 DOI: 10.1016/j.molcel.2024.01.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/07/2023] [Accepted: 01/25/2024] [Indexed: 02/23/2024]
Abstract
Cullin-RING ligases (CRLs) ubiquitylate specific substrates selected from other cellular proteins. Substrate discrimination and ubiquitin transferase activity were thought to be strictly separated. Substrates are recognized by substrate receptors, such as Fbox or BCbox proteins. Meanwhile, CRLs employ assorted ubiquitin-carrying enzymes (UCEs, which are a collection of E2 and ARIH-family E3s) specialized for either initial substrate ubiquitylation (priming) or forging poly-ubiquitin chains. We discovered specific human CRL-UCE pairings governing substrate priming. The results reveal pairing of CUL2-based CRLs and UBE2R-family UCEs in cells, essential for efficient PROTAC-induced neo-substrate degradation. Despite UBE2R2's intrinsic programming to catalyze poly-ubiquitylation, CUL2 employs this UCE for geometrically precise PROTAC-dependent ubiquitylation of a neo-substrate and for rapid priming of substrates recruited to diverse receptors. Cryo-EM structures illuminate how CUL2-based CRLs engage UBE2R2 to activate substrate ubiquitylation. Thus, pairing with a specific UCE overcomes E2 catalytic limitations to drive substrate ubiquitylation and targeted protein degradation.
Collapse
Affiliation(s)
- Jerry Li
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV 89154, USA
| | - Nicholas Purser
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV 89154, USA
| | - Joanna Liwocha
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Daniel C Scott
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Holly A Byers
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Barbara Steigenberger
- Mass Spectrometry Core Facility, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Spencer Hill
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV 89154, USA
| | - Ishita Tripathi-Giesgen
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Trent Hinkle
- Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Fynn M Hansen
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - J Rajan Prabu
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | | | | | | | - Brenda A Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany; Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Gary Kleiger
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV 89154, USA; Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried 82152, Germany.
| |
Collapse
|
6
|
Madunić K, Luijkx YMCA, Mayboroda OA, Janssen GMC, van Veelen PA, Strijbis K, Wennekes T, Lageveen-Kammeijer GSM, Wuhrer M. O-Glycomic and Proteomic Signatures of Spontaneous and Butyrate-Stimulated Colorectal Cancer Cell Line Differentiation. Mol Cell Proteomics 2023; 22:100501. [PMID: 36669592 PMCID: PMC9999233 DOI: 10.1016/j.mcpro.2023.100501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Gut microbiota of the gastrointestinal tract provide health benefits to the human host via bacterial metabolites. Bacterial butyrate has beneficial effects on intestinal homeostasis and is the preferred energy source of intestinal epithelial cells, capable of inducing differentiation. It was previously observed that changes in the expression of specific proteins as well as protein glycosylation occur with differentiation. In this study, specific mucin O-glycans were identified that mark butyrate-induced epithelial differentiation of the intestinal cell line CaCo-2 (Cancer Coli-2), by applying porous graphitized carbon nano-liquid chromatography with electrospray ionization tandem mass spectrometry. Moreover, a quantitative proteomic approach was used to decipher changes in the cell proteome. It was found that the fully differentiated butyrate-stimulated cells are characterized by a higher expression of sialylated O-glycan structures, whereas fucosylation is downregulated with differentiation. By performing an integrative approach, we generated hypotheses about the origin of the observed O-glycome changes. These insights pave the way for future endeavors to study the dynamic O-glycosylation patterns in the gut, either produced via cellular biosynthesis or through the action of bacterial glycosidases as well as the functional role of these patterns in homeostasis and dysbiosis at the gut-microbiota interface.
Collapse
Affiliation(s)
- K Madunić
- Center for Proteomics and Metabolomics, Leiden University, The Netherlands
| | - Y M C A Luijkx
- Department Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands; Department Biomolecular Health Sciences, Utrecht University, Utrecht, The Netherlands
| | - O A Mayboroda
- Center for Proteomics and Metabolomics, Leiden University, The Netherlands
| | - G M C Janssen
- Center for Proteomics and Metabolomics, Leiden University, The Netherlands
| | - P A van Veelen
- Center for Proteomics and Metabolomics, Leiden University, The Netherlands
| | - K Strijbis
- Department Biomolecular Health Sciences, Utrecht University, Utrecht, The Netherlands
| | - T Wennekes
- Department Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | | | - M Wuhrer
- Center for Proteomics and Metabolomics, Leiden University, The Netherlands.
| |
Collapse
|
7
|
Marei H, Tsai WTK, Kee YS, Ruiz K, He J, Cox C, Sun T, Penikalapati S, Dwivedi P, Choi M, Kan D, Saenz-Lopez P, Dorighi K, Zhang P, Kschonsak YT, Kljavin N, Amin D, Kim I, Mancini AG, Nguyen T, Wang C, Janezic E, Doan A, Mai E, Xi H, Gu C, Heinlein M, Biehs B, Wu J, Lehoux I, Harris S, Comps-Agrar L, Seshasayee D, de Sauvage FJ, Grimmer M, Li J, Agard NJ, de Sousa E Melo F. Antibody targeting of E3 ubiquitin ligases for receptor degradation. Nature 2022; 610:182-189. [PMID: 36131013 PMCID: PMC9534761 DOI: 10.1038/s41586-022-05235-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 08/12/2022] [Indexed: 12/31/2022]
Abstract
Most current therapies that target plasma membrane receptors function by antagonizing ligand binding or enzymatic activities. However, typical mammalian proteins comprise multiple domains that execute discrete but coordinated activities. Thus, inhibition of one domain often incompletely suppresses the function of a protein. Indeed, targeted protein degradation technologies, including proteolysis-targeting chimeras1 (PROTACs), have highlighted clinically important advantages of target degradation over inhibition2. However, the generation of heterobifunctional compounds binding to two targets with high affinity is complex, particularly when oral bioavailability is required3. Here we describe the development of proteolysis-targeting antibodies (PROTABs) that tether cell-surface E3 ubiquitin ligases to transmembrane proteins, resulting in target degradation both in vitro and in vivo. Focusing on zinc- and ring finger 3 (ZNRF3), a Wnt-responsive ligase, we show that this approach can enable colorectal cancer-specific degradation. Notably, by examining a matrix of additional cell-surface E3 ubiquitin ligases and transmembrane receptors, we demonstrate that this technology is amendable for ‘on-demand’ degradation. Furthermore, we offer insights on the ground rules governing target degradation by engineering optimized antibody formats. In summary, this work describes a strategy for the rapid development of potent, bioavailable and tissue-selective degraders of cell-surface proteins. Membrane-bound E3 ubiquitin ligases RNF43 and ZNRF3 are overexpressed in colorectal cancer, and can be repurposed using proteolysis-targeting antibodies (PROTABs) to selectively degrade cell-surface receptors in tumours.
Collapse
Affiliation(s)
- Hadir Marei
- Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Wen-Ting K Tsai
- Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Yee-Seir Kee
- Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Karen Ruiz
- Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Jieyan He
- Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - Chris Cox
- Discovery Immunology, Genentech Inc, South San Francisco, CA, USA
| | - Tao Sun
- Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Sai Penikalapati
- Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, CA, USA
| | - Pankaj Dwivedi
- Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, CA, USA
| | - Meena Choi
- Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, CA, USA
| | - David Kan
- Translational Oncology, Genentech, South San Francisco, CA, USA
| | | | | | - Pamela Zhang
- Antibody Engineering, Genentech, South San Francisco, CA, USA
| | | | - Noelyn Kljavin
- Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Dhara Amin
- Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Ingrid Kim
- Antibody Engineering, Genentech, South San Francisco, CA, USA
| | | | - Thao Nguyen
- Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Chunling Wang
- Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Eric Janezic
- Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - Alexander Doan
- Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - Elaine Mai
- Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - Hongkang Xi
- Antibody discovery, Genentech, South San Francisco, CA, USA
| | - Chen Gu
- Protein Chemistry, Genentech, South San Francisco, CA, USA
| | | | - Brian Biehs
- Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Jia Wu
- Antibody discovery, Genentech, South San Francisco, CA, USA
| | - Isabelle Lehoux
- Biomolecular Resources, Genentech, South San Francisco, CA, USA
| | - Seth Harris
- Structural Biology, Genentech, South San Francisco, CA, USA
| | | | | | | | | | - Jing Li
- Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | | | | |
Collapse
|
8
|
Rossi M, Altea-Manzano P, Demicco M, Doglioni G, Bornes L, Fukano M, Vandekeere A, Cuadros AM, Fernández-García J, Riera-Domingo C, Jauset C, Planque M, Alkan HF, Nittner D, Zuo D, Broadfield LA, Parik S, Pane AA, Rizzollo F, Rinaldi G, Zhang T, Teoh ST, Aurora AB, Karras P, Vermeire I, Broekaert D, Elsen JV, Knott MML, Orth MF, Demeyer S, Eelen G, Dobrolecki LE, Bassez A, Brussel TV, Sotlar K, Lewis MT, Bartsch H, Wuhrer M, Veelen PV, Carmeliet P, Cools J, Morrison SJ, Marine JC, Lambrechts D, Mazzone M, Hannon GJ, Lunt SY, Grünewald TGP, Park M, Rheenen JV, Fendt SM. PHGDH heterogeneity potentiates cancer cell dissemination and metastasis. Nature 2022; 605:747-753. [PMID: 35585241 PMCID: PMC9888363 DOI: 10.1038/s41586-022-04758-2] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 04/12/2022] [Indexed: 02/02/2023]
Abstract
Cancer metastasis requires the transient activation of cellular programs enabling dissemination and seeding in distant organs1. Genetic, transcriptional and translational heterogeneity contributes to this dynamic process2,3. Metabolic heterogeneity has also been observed4, yet its role in cancer progression is less explored. Here we find that the loss of phosphoglycerate dehydrogenase (PHGDH) potentiates metastatic dissemination. Specifically, we find that heterogeneous or low PHGDH expression in primary tumours of patients with breast cancer is associated with decreased metastasis-free survival time. In mice, circulating tumour cells and early metastatic lesions are enriched with Phgdhlow cancer cells, and silencing Phgdh in primary tumours increases metastasis formation. Mechanistically, Phgdh interacts with the glycolytic enzyme phosphofructokinase, and the loss of this interaction activates the hexosamine-sialic acid pathway, which provides precursors for protein glycosylation. As a consequence, aberrant protein glycosylation occurs, including increased sialylation of integrin αvβ3, which potentiates cell migration and invasion. Inhibition of sialylation counteracts the metastatic ability of Phgdhlow cancer cells. In conclusion, although the catalytic activity of PHGDH supports cancer cell proliferation, low PHGDH protein expression non-catalytically potentiates cancer dissemination and metastasis formation. Thus, the presence of PHDGH heterogeneity in primary tumours could be considered a sign of tumour aggressiveness.
Collapse
Affiliation(s)
- Matteo Rossi
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Patricia Altea-Manzano
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Margherita Demicco
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Ginevra Doglioni
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Laura Bornes
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marina Fukano
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
- Rosalind & Morris Goodman Cancer Institute (GCI), McGill University, Montreal, Quebec, Canada
| | - Anke Vandekeere
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Alejandro M Cuadros
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Juan Fernández-García
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Carla Riera-Domingo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Cristina Jauset
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Mélanie Planque
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - H Furkan Alkan
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - David Nittner
- Histopathology Expertise Center, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Dongmei Zuo
- Rosalind & Morris Goodman Cancer Institute (GCI), McGill University, Montreal, Quebec, Canada
| | - Lindsay A Broadfield
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Sweta Parik
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Antonino Alejandro Pane
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Francesca Rizzollo
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Gianmarco Rinaldi
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Shao Thing Teoh
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Arin B Aurora
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Panagiotis Karras
- Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
| | - Ines Vermeire
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Dorien Broekaert
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Joke Van Elsen
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Maximilian M L Knott
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Martin F Orth
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sofie Demeyer
- Laboratory for Molecular Biology of Leukemia, VIB-KU Leuven, Leuven, Belgium
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | | | - Ayse Bassez
- Laboratory for Translational Genetics, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Thomas Van Brussel
- Laboratory for Translational Genetics, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Karl Sotlar
- Institute of Pathology, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | | | - Harald Bartsch
- Institute of Pathology, Ludwig Maximilians University, Munich, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jan Cools
- Laboratory for Molecular Biology of Leukemia, VIB-KU Leuven, Leuven, Belgium
| | - Sean J Morrison
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jean-Christophe Marine
- Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Molecular Biotechnology and Health Science, Molecular Biotechnology Centre, University of Torino, Torino, Italy
| | - Gregory J Hannon
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, USA
| | - Thomas G P Grünewald
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Morag Park
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
- Rosalind & Morris Goodman Cancer Institute (GCI), McGill University, Montreal, Quebec, Canada
| | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium.
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium.
| |
Collapse
|
9
|
Mirhadi S, Tam S, Li Q, Moghal N, Pham NA, Tong J, Golbourn BJ, Krieger JR, Taylor P, Li M, Weiss J, Martins-Filho SN, Raghavan V, Mamatjan Y, Khan AA, Cabanero M, Sakashita S, Huo K, Agnihotri S, Ishizawa K, Waddell TK, Zadeh G, Yasufuku K, Liu G, Shepherd FA, Moran MF, Tsao MS. Integrative analysis of non-small cell lung cancer patient-derived xenografts identifies distinct proteotypes associated with patient outcomes. Nat Commun 2022; 13:1811. [PMID: 35383171 PMCID: PMC8983714 DOI: 10.1038/s41467-022-29444-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/17/2022] [Indexed: 12/24/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer deaths worldwide. Only a fraction of NSCLC harbor actionable driver mutations and there is an urgent need for patient-derived model systems that will enable the development of new targeted therapies. NSCLC and other cancers display profound proteome remodeling compared to normal tissue that is not predicted by DNA or RNA analyses. Here, we generate 137 NSCLC patient-derived xenografts (PDXs) that recapitulate the histology and molecular features of primary NSCLC. Proteome analysis of the PDX models reveals 3 adenocarcinoma and 2 squamous cell carcinoma proteotypes that are associated with different patient outcomes, protein-phosphotyrosine profiles, signatures of activated pathways and candidate targets, and in adenocarcinoma, stromal immune features. These findings portend proteome-based NSCLC classification and treatment and support the PDX resource as a viable model for the development of new targeted therapies. With non-small cell lung cancer (NSCLC) being the leading cause of cancer deaths worldwide, the development of targeted therapies remains crucial. Here, the generation and multi-omics characterization of 137 NSCLC patient-derived xenografts provides a resource for potential classifications and targets.
Collapse
Affiliation(s)
- Shideh Mirhadi
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Shirley Tam
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Quan Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Nadeem Moghal
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Nhu-An Pham
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jiefei Tong
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Brian J Golbourn
- John G. Rangos Sr. Research Center, Children's Hospital of Pittsburgh, and Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Paul Taylor
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Ming Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jessica Weiss
- Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Sebastiao N Martins-Filho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Vibha Raghavan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yasin Mamatjan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Aafaque A Khan
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Michael Cabanero
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Shingo Sakashita
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Kugeng Huo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sameer Agnihotri
- John G. Rangos Sr. Research Center, Children's Hospital of Pittsburgh, and Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kota Ishizawa
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Thomas K Waddell
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Gelareh Zadeh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Kazuhiro Yasufuku
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Frances A Shepherd
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, ON, Canada
| | - Michael F Moran
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada. .,Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
10
|
Budayeva HG, Sengupta-Ghosh A, Phu L, Moffat JG, Ayalon G, Kirkpatrick DS. Phosphoproteome Profiling of the Receptor Tyrosine Kinase MuSK Identifies Tyrosine Phosphorylation of Rab GTPases. Mol Cell Proteomics 2022; 21:100221. [PMID: 35227894 PMCID: PMC8972003 DOI: 10.1016/j.mcpro.2022.100221] [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: 06/23/2021] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/16/2022] Open
Abstract
Muscle-specific receptor tyrosine kinase (MuSK) agonist antibodies were developed 2 decades ago to explore the benefits of receptor activation at the neuromuscular junction. Unlike agrin, the endogenous agonist of MuSK, agonist antibodies function independently of its coreceptor low-density lipoprotein receptor–related protein 4 to delay the onset of muscle denervation in mouse models of ALS. Here, we performed dose–response and time-course experiments on myotubes to systematically compare site-specific phosphorylation downstream of each agonist. Remarkably, both agonists elicited similar intracellular responses at known and newly identified MuSK signaling components. Among these was inducible tyrosine phosphorylation of multiple Rab GTPases that was blocked by MuSK inhibition. Importantly, mutation of this site in Rab10 disrupts association with its effector proteins, molecule interacting with CasL 1/3. Together, these data provide in-depth characterization of MuSK signaling, describe two novel MuSK inhibitors, and expose phosphorylation of Rab GTPases downstream of receptor tyrosine kinase activation in myotubes. Different agonists of muscle-specific kinase (MuSK) elicit similar phosphoprofiles. MuSK activation induces tyrosine phosphorylation of several Rab GTPases. MuSK inhibitors diminish receptor signaling, including phosphorylation on Rab10 Y6. Mutation of Rab10 Y6 disrupts its association with Mical adaptor proteins.
Collapse
Affiliation(s)
- Hanna G Budayeva
- Microchemistry, Proteomics, and Lipidomics Department, Genentech, Inc, South San Francisco, California, USA.
| | | | - Lilian Phu
- Microchemistry, Proteomics, and Lipidomics Department, Genentech, Inc, South San Francisco, California, USA
| | - John G Moffat
- Biochemical and Cellular Pharmacology and Computational Drug Design, Genentech, Inc, South San Francisco, California, USA
| | - Gai Ayalon
- Neuroscience Department, Genentech, Inc, South San Francisco, California, USA
| | - Donald S Kirkpatrick
- Microchemistry, Proteomics, and Lipidomics Department, Genentech, Inc, South San Francisco, California, USA.
| |
Collapse
|
11
|
Zmyslowska A, Kuljanin M, Malachowska B, Stanczak M, Michalek D, Wlodarczyk A, Grot D, Taha J, Pawlik B, Lebiedzińska-Arciszewska M, Nieznanska H, Wieckowski MR, Rieske P, Mancias JD, Borowiec M, Mlynarski W, Fendler W. Multiomic analysis on human cell model of wolfram syndrome reveals changes in mitochondrial morphology and function. Cell Commun Signal 2021; 19:116. [PMID: 34801048 PMCID: PMC8605533 DOI: 10.1186/s12964-021-00791-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/01/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Wolfram syndrome (WFS) is a rare autosomal recessive syndrome in which diabetes mellitus and neurodegenerative disorders occur as a result of Wolframin deficiency and increased ER stress. In addition, WFS1 deficiency leads to calcium homeostasis disturbances and can change mitochondrial dynamics. The aim of this study was to evaluate protein levels and changes in gene transcription on human WFS cell model under experimental ER stress. METHODS We performed transcriptomic and proteomic analysis on WFS human cell model-skin fibroblasts reprogrammed into induced pluripotent stem (iPS) cells and then into neural stem cells (NSC) with subsequent ER stress induction using tunicamycin (TM). Results were cross-referenced with publicly available RNA sequencing data in hippocampi and hypothalami of mice with WFS1 deficiency. RESULTS Proteomic analysis identified specific signal pathways that differ in NSC WFS cells from healthy ones. Next, detailed analysis of the proteins involved in the mitochondrial function showed the down-regulation of subunits of the respiratory chain complexes in NSC WFS cells, as well as the up-regulation of proteins involved in Krebs cycle and glycolysis when compared to the control cells. Based on pathway enrichment analysis we concluded that in samples from mice hippocampi the mitochondrial protein import machinery and OXPHOS were significantly down-regulated. CONCLUSIONS Our results show the functional and morphological secondary mitochondrial damage in patients with WFS. Video Abstract.
Collapse
Affiliation(s)
- Agnieszka Zmyslowska
- Department of Clinical Genetics, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland
| | - Miljan Kuljanin
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Beata Malachowska
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
- Department of Radiation Oncology, Einstein College of Medicine, Bronx, NY USA
| | - Marcin Stanczak
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Dominika Michalek
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Aneta Wlodarczyk
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
| | - Dagmara Grot
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
| | - Joanna Taha
- Central Laboratory for Genetic Research in Pediatric Oncology “Oncolab”, Medical University of Lodz, Lodz, Poland
| | - Bartłomiej Pawlik
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | - Hanna Nieznanska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Mariusz R. Wieckowski
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Rieske
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
| | - Joseph D. Mancias
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Maciej Borowiec
- Department of Clinical Genetics, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland
| | - Wojciech Mlynarski
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
| | - Wojciech Fendler
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
12
|
Liu X, Gygi SP, Paulo JA. A Semiautomated Paramagnetic Bead-Based Platform for Isobaric Tag Sample Preparation. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:1519-1529. [PMID: 33950666 PMCID: PMC8210952 DOI: 10.1021/jasms.1c00077] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The development of streamlined and high-throughput sample processing workflows is important for capitalizing on emerging advances and innovations in mass spectrometry-based applications. While the adaptation of new technologies and improved methodologies is fast paced, automation of upstream sample processing often lags. Here we have developed and implemented a semiautomated paramagnetic bead-based platform for isobaric tag sample preparation. We benchmarked the robot-assisted platform by comparing the protein abundance profiles of six common parental laboratory yeast strains in triplicate TMTpro16-plex experiments against an identical set of experiments in which the samples were manually processed. Both sets of experiments quantified similar numbers of proteins and peptides with good reproducibility. Using these data, we constructed an interactive website to explore the proteome profiles of six yeast strains. We also provide the community with open-source templates for automating routine proteomics workflows on an opentrons OT-2 liquid handler. The robot-assisted platform offers a versatile and affordable option for reproducible sample processing for a wide range of protein profiling applications.
Collapse
Affiliation(s)
- Xinyue Liu
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
13
|
Temporal proteomic changes induced by nicotine in human cells: A quantitative proteomics approach. J Proteomics 2021; 241:104244. [PMID: 33895337 DOI: 10.1016/j.jprot.2021.104244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/04/2021] [Accepted: 04/19/2021] [Indexed: 12/20/2022]
Abstract
Nicotine is a prominent active compound in tobacco and many smoking cessation products. Some of the biological effects of nicotine are well documented in in vitro and in vivo systems; however, data are scarce concerning the time-dependent changes on protein and phosphorylation events in response to nicotine. Here, we profiled the proteomes of SH-SY5Y and A549 cell lines subjected to acute (15 min, 1 h and 4 h) or chronic (24 h, 48 h) nicotine exposures. We used sample multiplexing (TMTpro16) and quantified more than 9000 proteins and over 7000 phosphorylation events per cell line. Among our findings, we determined a decrease in mitochondrial protein abundance for SH-SY5Y, while we detected alterations in several immune pathways, such as the complement system, for A549 following nicotine treatment. We also explored the proposed association between smoking (specifically nicotine) and SARS-CoV2. Here, we found several host proteins known to interact with viral proteins that were affected by nicotine in a time dependent manner. This dataset can be mined further to investigate the potential role of nicotine in different biological contexts. SIGNIFICANCE: Smoking is a major public health issue that is associated with several serious chronic, yet preventable diseases, including stroke, heart disease, type 2 diabetes, cancer, and susceptibility to infection. Tobacco smoke is a complex mixture of thousands of different compounds, among which nicotine is the main addictive compound. The biological effects of nicotine have been reported in several models, however very little data are available concerning the temporal proteomic and phosphoproteomic changes in response to nicotine. Here, we provide a dataset exploring the potential role of nicotine on different biological processes over time, including implications in the study of SARS-CoV2.
Collapse
|
14
|
Malone CF, Dharia NV, Kugener G, Forman AB, Rothberg MV, Abdusamad M, Gonzalez A, Kuljanin M, Robichaud AL, Conway AS, Dempster JM, Paolella BR, Dumont N, Hovestadt V, Mancias JD, Younger ST, Root DE, Golub TR, Vazquez F, Stegmaier K. Selective Modulation of a Pan-Essential Protein as a Therapeutic Strategy in Cancer. Cancer Discov 2021; 11:2282-2299. [PMID: 33883167 DOI: 10.1158/2159-8290.cd-20-1213] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 02/12/2021] [Accepted: 03/26/2021] [Indexed: 12/26/2022]
Abstract
Cancer dependency maps, which use CRISPR/Cas9 depletion screens to profile the landscape of genetic dependencies in hundreds of cancer cell lines, have identified context-specific dependencies that could be therapeutically exploited. An ideal therapy is both lethal and precise, but these depletion screens cannot readily distinguish between gene effects that are cytostatic or cytotoxic. Here, we use a diverse panel of functional genomic screening assays to identify NXT1 as a selective and rapidly lethal in vivo relevant genetic dependency in MYCN-amplified neuroblastoma. NXT1 heterodimerizes with NXF1, and together they form the principal mRNA nuclear export machinery. We describe a previously unrecognized mechanism of synthetic lethality between NXT1 and its paralog NXT2: their common essential binding partner NXF1 is lost only in the absence of both. We propose a potential therapeutic strategy for tumor-selective elimination of a protein that, if targeted directly, is expected to cause widespread toxicity. SIGNIFICANCE: We provide a framework for identifying new therapeutic targets from functional genomic screens. We nominate NXT1 as a selective lethal target in neuroblastoma and propose a therapeutic approach where the essential protein NXF1 can be selectively eliminated in tumor cells by exploiting the NXT1-NXT2 paralog relationship.See related commentary by Wang and Abdel-Wahab, p. 2129.This article is highlighted in the In This Issue feature, p. 2113.
Collapse
Affiliation(s)
- Clare F Malone
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Neekesh V Dharia
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | | | - Alexandra B Forman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | | | - Mai Abdusamad
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | - Miljan Kuljanin
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amanda L Robichaud
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amy Saur Conway
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | - Nancy Dumont
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Volker Hovestadt
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | - Joseph D Mancias
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Scott T Younger
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - David E Root
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Todd R Golub
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | | | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
15
|
Navarrete-Perea J, Gygi SP, Paulo JA. HYpro16: A Two-Proteome Mixture to Assess Interference in Isobaric Tag-Based Sample Multiplexing Experiments. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:247-254. [PMID: 33175540 PMCID: PMC8210950 DOI: 10.1021/jasms.0c00299] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Isobaric tagging is a powerful strategy for global proteome profiling. A caveat of isobaric-tag-based quantification is "interference", which may be caused by coeluting peptides that are coisolated, cofragmented, and coanalyzed, thereby confounding quantitative accuracy. Here, we present a two-proteome standard that challenges the mass spectrometer to measure a range of protein abundance ratios in a background of potential interference. The HYpro16 standard consists of tandem mass tag (TMT) pro16-labeled human peptides at a 1:1 ratio across all channels into which is spiked TMTpro16-labeled yeast peptides in triplicate at 20:1, 10:1, 4:1, and 2:1 ratios. We showcase the HYpro16 standard by (1) altering the MS2 isolation window width and (2) examining different data acquisition methods (hrMS2, SPS-MS3, RTS-MS3). Our data illustrate that wider isolation widths moderately increase the TMT signal, the benefits of which are offset by decreased ratio accuracy. We also show that using real-time database searching (RTS)-MS3 resulted in the most accurate ratios. Additionally, the number of quantified yeast proteins using RTS-MS3 approaches that of hrMS2 when using a yeast-specific database for real-time searching. In short, this quality control standard allows for the assessment of multiple quantitative measurements within a single run, which can be compared across instruments to benchmark and track performance.
Collapse
Affiliation(s)
- Jose Navarrete-Perea
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
16
|
Banh RS, Biancur DE, Yamamoto K, Sohn ASW, Walters B, Kuljanin M, Gikandi A, Wang H, Mancias JD, Schneider RJ, Pacold ME, Kimmelman AC. Neurons Release Serine to Support mRNA Translation in Pancreatic Cancer. Cell 2020; 183:1202-1218.e25. [PMID: 33142117 DOI: 10.1016/j.cell.2020.10.016] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/31/2020] [Accepted: 10/06/2020] [Indexed: 12/23/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) tumors have a nutrient-poor, desmoplastic, and highly innervated tumor microenvironment. Although neurons can release stimulatory factors to accelerate PDAC tumorigenesis, the metabolic contribution of peripheral axons has not been explored. We found that peripheral axons release serine (Ser) to support the growth of exogenous Ser (exSer)-dependent PDAC cells during Ser/Gly (glycine) deprivation. Ser deprivation resulted in ribosomal stalling on two of the six Ser codons, TCC and TCT, and allowed the selective translation and secretion of nerve growth factor (NGF) by PDAC cells to promote tumor innervation. Consistent with this, exSer-dependent PDAC tumors grew slower and displayed enhanced innervation in mice on a Ser/Gly-free diet. Blockade of compensatory neuronal innervation using LOXO-101, a Trk-NGF inhibitor, further decreased PDAC tumor growth. Our data indicate that axonal-cancer metabolic crosstalk is a critical adaptation to support PDAC growth in nutrient poor environments.
Collapse
Affiliation(s)
- Robert S Banh
- Department of Radiation Oncology, Perlmutter Cancer Center, New York University Medical Center, New York, NY 10016, USA
| | - Douglas E Biancur
- Department of Radiation Oncology, Perlmutter Cancer Center, New York University Medical Center, New York, NY 10016, USA
| | - Keisuke Yamamoto
- Department of Radiation Oncology, Perlmutter Cancer Center, New York University Medical Center, New York, NY 10016, USA
| | - Albert S W Sohn
- Department of Radiation Oncology, Perlmutter Cancer Center, New York University Medical Center, New York, NY 10016, USA
| | - Beth Walters
- Department of Microbiology and Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - Miljan Kuljanin
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Hale Family Pancreatic Cancer Research Center, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ajami Gikandi
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Hale Family Pancreatic Cancer Research Center, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Huamin Wang
- Department of Anatomical Pathology, M.D. Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Joseph D Mancias
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Hale Family Pancreatic Cancer Research Center, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Robert J Schneider
- Department of Radiation Oncology, Perlmutter Cancer Center, New York University Medical Center, New York, NY 10016, USA; Department of Microbiology and Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - Michael E Pacold
- Department of Radiation Oncology, Perlmutter Cancer Center, New York University Medical Center, New York, NY 10016, USA
| | - Alec C Kimmelman
- Department of Radiation Oncology, Perlmutter Cancer Center, New York University Medical Center, New York, NY 10016, USA.
| |
Collapse
|
17
|
Zhang T, Gygi SP, Paulo JA. Temporal Proteomic Profiling of SH-SY5Y Differentiation with Retinoic Acid Using FAIMS and Real-Time Searching. J Proteome Res 2020; 20:704-714. [PMID: 33054241 DOI: 10.1021/acs.jproteome.0c00614] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The SH-SY5Y cell line is often used as a surrogate for neurons in cell-based studies. This cell line is frequently differentiated with all-trans retinoic acid (ATRA) over a 7-day period, which confers neuron-like properties to the cells. However, no analysis of proteome remodeling has followed the progress of this transition. Here, we quantitatively profiled over 9400 proteins across a 7-day treatment with retinoic acid using state-of-the-art mass spectrometry-based proteomics technologies, including FAIMS, real-time database searching, and TMTpro16 sample multiplexing. Gene ontology analysis revealed that categories with the highest increases in protein abundance were related to the plasma membrane/extracellular space. To showcase our data set, we surveyed the protein abundance profiles linked to neurofilament bundle assembly, neuron projections, and neuronal cell body formation. These proteins exhibited increases in abundance level, yet we observed multiple patterns among the queried proteins. The data presented represent a rich resource for investigating temporal protein abundance changes in SH-SY5Y cells differentiated with retinoic acid. Moreover, the sample preparation and data acquisition strategies used here can be readily applied to any analogous cell line differentiation analysis.
Collapse
Affiliation(s)
- Tian Zhang
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, Massachusetts 02115, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, Massachusetts 02115, United States
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, Massachusetts 02115, United States
| |
Collapse
|
18
|
Navarrete-Perea J, Gygi SP, Paulo JA. Growth media selection alters the proteome profiles of three model microorganisms. J Proteomics 2020; 231:104006. [PMID: 33038513 DOI: 10.1016/j.jprot.2020.104006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/26/2020] [Accepted: 10/01/2020] [Indexed: 12/21/2022]
Abstract
The selection of growth media is a very important consideration of any cell-based proteomics experiment. Alterations thereof may result in differences in basal proteomes simply due to disparities in the metabolite composition of the media. We investigate the effect of growth media on the proteomes of three microorganisms, specifically E. coli, S. cerevisiae, and S. pombe, using tandem mass tag (TMT)-based quantitative proteomics. We compared the protein abundance profiles of these microorganisms propagated in two distinct growth media that are commonly used for the respective organism. Our sample preparation strategy included SP3 bead-assisted protein isolation and digestion. In addition, we assembled a replicate set of samples in which we altered the proteolytic digestion from sequential treatment with LysC and trypsin to only LysC. Despite differences in peptides identified and a drop in quantified proteins, the results were similar between the two datasets for all three microorganisms. Approximately 10% of the proteins of each respective microorganism were significantly altered in each dataset. As expected, gene ontology analysis revealed that the majority of differentially expressed proteins are implicated in metabolism. These data emphasize further the importance and the potential consequences of growth media selection. SIGNIFICANCE: Various microorganisms are used as model systems throughout in biological studies, including proteomics-based investigations. The growth conditions of these organisms are of utmost importance, of which one major consideration is the choice of growth media. We hypothesize that growth media selection has a considerable impact on the baseline proteome of a given microorganism. To test this hypothesis, we used tandem mass tag (TMT)-based quantitative multiplexed proteomics to profile the proteomes of E. coli, S. cerevisiae, and S. pombe each grown in two different, yet common, growth media for the respective species. Our data show that approximately 10% of the proteins of each respective microorganism were significantly altered and that many of the differentially expressed proteins are implicated in metabolism. We provide several datasets which are potentially valuable for growth media selection with respect to downstream biochemical analysis.
Collapse
Affiliation(s)
- Jose Navarrete-Perea
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
19
|
Nabet B, Ferguson FM, Seong BKA, Kuljanin M, Leggett AL, Mohardt ML, Robichaud A, Conway AS, Buckley DL, Mancias JD, Bradner JE, Stegmaier K, Gray NS. Rapid and direct control of target protein levels with VHL-recruiting dTAG molecules. Nat Commun 2020; 11:4687. [PMID: 32948771 PMCID: PMC7501296 DOI: 10.1038/s41467-020-18377-w] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Chemical biology strategies for directly perturbing protein homeostasis including the degradation tag (dTAG) system provide temporal advantages over genetic approaches and improved selectivity over small molecule inhibitors. We describe dTAGV-1, an exclusively selective VHL-recruiting dTAG molecule, to rapidly degrade FKBP12F36V-tagged proteins. dTAGV-1 overcomes a limitation of previously reported CRBN-recruiting dTAG molecules to degrade recalcitrant oncogenes, supports combination degrader studies and facilitates investigations of protein function in cells and mice.
Collapse
Affiliation(s)
- Behnam Nabet
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| | - Fleur M Ferguson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Bo Kyung A Seong
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Miljan Kuljanin
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alan L Leggett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mikaela L Mohardt
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amanda Robichaud
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amy S Conway
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Dennis L Buckley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Joseph D Mancias
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Abstract
Serine hydroxymethyltransferase 2 (SHMT2) converts serine plus tetrahydrofolate (THF) into glycine plus methylene-THF and is upregulated at the protein level in lung and other cancers. In order to better understand the role of SHMT2 in cancer a model system of HeLa cells engineered for inducible over-expression or knock-down of SHMT2 was characterized for cell proliferation and changes in metabolites and proteome as a function of SHMT2. Ectopic over-expression of SHMT2 increased cell proliferation in vitro and tumor growth in vivo. Knockdown of SHMT2 expression in vitro caused a state of glycine auxotrophy and accumulation of phosphoribosylaminoimidazolecarboxamide (AICAR), an intermediate of folate/1-carbon-pathway-dependent de novo purine nucleotide synthesis. Decreased glycine in the HeLa cell-based xenograft tumors with knocked down SHMT2 was potentiated by administration of the anti-hyperglycinemia agent benzoate. However, tumor growth was not affected by SHMT2 knockdown with or without benzoate treatment. Benzoate inhibited cell proliferation in vitro, but this was independent of SHMT2 modulation. The abundance of proteins of mitochondrial respiration complexes 1 and 3 was inversely correlated with SHMT2 levels. Proximity biotinylation in vivo (BioID) identified 48 mostly mitochondrial proteins associated with SHMT2 including the mitochondrial enzymes Acyl-CoA thioesterase (ACOT2) and glutamate dehydrogenase (GLUD1) along with more than 20 proteins from mitochondrial respiration complexes 1 and 3. These data provide insights into possible mechanisms through which elevated SHMT2 in cancers may be linked to changes in metabolism and mitochondrial function.
Collapse
|
21
|
Nowinski SM, Solmonson A, Rusin SF, Maschek JA, Bensard CL, Fogarty S, Jeong MY, Lettlova S, Berg JA, Morgan JT, Ouyang Y, Naylor BC, Paulo JA, Funai K, Cox JE, Gygi SP, Winge DR, DeBerardinis RJ, Rutter J. Mitochondrial fatty acid synthesis coordinates oxidative metabolism in mammalian mitochondria. eLife 2020; 9:58041. [PMID: 32804083 PMCID: PMC7470841 DOI: 10.7554/elife.58041] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/15/2020] [Indexed: 12/13/2022] Open
Abstract
Cells harbor two systems for fatty acid synthesis, one in the cytoplasm (catalyzed by fatty acid synthase, FASN) and one in the mitochondria (mtFAS). In contrast to FASN, mtFAS is poorly characterized, especially in higher eukaryotes, with the major product(s), metabolic roles, and cellular function(s) being essentially unknown. Here we show that hypomorphic mtFAS mutant mouse skeletal myoblast cell lines display a severe loss of electron transport chain (ETC) complexes and exhibit compensatory metabolic activities including reductive carboxylation. This effect on ETC complexes appears to be independent of protein lipoylation, the best characterized function of mtFAS, as mutants lacking lipoylation have an intact ETC. Finally, mtFAS impairment blocks the differentiation of skeletal myoblasts in vitro. Together, these data suggest that ETC activity in mammals is profoundly controlled by mtFAS function, thereby connecting anabolic fatty acid synthesis with the oxidation of carbon fuels.
Collapse
Affiliation(s)
| | - Ashley Solmonson
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Scott F Rusin
- Department of Cell Biology, Harvard University School of Medicine, Boston, United States
| | - J Alan Maschek
- Diabetes & Metabolism Research Center, Salt Lake City, United States.,Department of Nutrition and Integrative Physiology, Salt Lake City, United States.,Metabolomics, Proteomics and Mass Spectrometry Core Research Facilities University of Utah, Salt Lake City, United States
| | | | - Sarah Fogarty
- Department of Biochemistry, Salt Lake City, United States.,Howard Hughes Medical Institute, Salt Lake City, United States
| | - Mi-Young Jeong
- Department of Biochemistry, Salt Lake City, United States
| | | | - Jordan A Berg
- Department of Biochemistry, Salt Lake City, United States
| | - Jeffrey T Morgan
- Department of Biochemistry, Salt Lake City, United States.,Howard Hughes Medical Institute, Salt Lake City, United States
| | - Yeyun Ouyang
- Department of Biochemistry, Salt Lake City, United States
| | - Bradley C Naylor
- Metabolomics, Proteomics and Mass Spectrometry Core Research Facilities University of Utah, Salt Lake City, United States
| | - Joao A Paulo
- Department of Cell Biology, Harvard University School of Medicine, Boston, United States
| | - Katsuhiko Funai
- Diabetes & Metabolism Research Center, Salt Lake City, United States
| | - James E Cox
- Department of Biochemistry, Salt Lake City, United States.,Diabetes & Metabolism Research Center, Salt Lake City, United States.,Metabolomics, Proteomics and Mass Spectrometry Core Research Facilities University of Utah, Salt Lake City, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard University School of Medicine, Boston, United States
| | - Dennis R Winge
- Department of Biochemistry, Salt Lake City, United States.,Diabetes & Metabolism Research Center, Salt Lake City, United States.,Department of Internal Medicine, Salt Lake City, United States
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, United States.,Howard Hughes Medical Institute, Salt Lake City, United States
| | - Jared Rutter
- Department of Biochemistry, Salt Lake City, United States.,Diabetes & Metabolism Research Center, Salt Lake City, United States.,Howard Hughes Medical Institute, Salt Lake City, United States
| |
Collapse
|
22
|
N-Glycoproteins Have a Major Role in MGL Binding to Colorectal Cancer Cell Lines: Associations with Overall Proteome Diversity. Int J Mol Sci 2020; 21:ijms21155522. [PMID: 32752259 PMCID: PMC7432225 DOI: 10.3390/ijms21155522] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 12/25/2022] Open
Abstract
Colorectal cancer (CRC) is the second-leading cause of cancer death worldwide due in part to a high proportion of patients diagnosed at advanced stages of the disease. For this reason, many efforts have been made towards new approaches for early detection and prognosis. Cancer-associated aberrant glycosylation, especially the Tn and STn antigens, can be detected using the macrophage galactose-type C-type lectin (MGL/CLEC10A/CD301), which has been shown to be a promising tool for CRC prognosis. We had recently identified the major MGL-binding glycoproteins in two high-MGL-binding CRC cells lines, HCT116 and HT29. However, we failed to detect the presence of O-linked Tn and STn glycans on most CRC glycoproteins recognized by MGL. We therefore investigated here the impact of N-linked and O-linked glycans carried by these proteins for the binding to MGL. In addition, we performed quantitative proteomics to study the major differences in proteins involved in glycosylation in these cells. Our results showed that N-glycans have a significant, previously underestimated, importance in MGL binding to CRC cell lines. Finally, we highlighted both common and cell-specific processes associated with a high-MGL-binding phenotype, such as differential levels of enzymes involved in protein glycosylation, and a transcriptional factor (CDX-2) involved in their regulation.
Collapse
|
23
|
Ferguson FM, Nabet B, Raghavan S, Liu Y, Leggett AL, Kuljanin M, Kalekar RL, Yang A, He S, Wang J, Ng RWS, Sulahian R, Li L, Poulin EJ, Huang L, Koren J, Dieguez-Martinez N, Espinosa S, Zeng Z, Corona CR, Vasta JD, Ohi R, Sim T, Kim ND, Harshbarger W, Lizcano JM, Robers MB, Muthaswamy S, Lin CY, Look AT, Haigis KM, Mancias JD, Wolpin BM, Aguirre AJ, Hahn WC, Westover KD, Gray NS. Discovery of a selective inhibitor of doublecortin like kinase 1. Nat Chem Biol 2020; 16:635-643. [PMID: 32251410 PMCID: PMC7246176 DOI: 10.1038/s41589-020-0506-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/05/2020] [Accepted: 02/24/2020] [Indexed: 12/16/2022]
Abstract
Doublecortin like kinase 1 (DCLK1) is an understudied kinase that is upregulated in a wide range of cancers, including pancreatic ductal adenocarcinoma (PDAC). However, little is known about its potential as a therapeutic target. We used chemoproteomic profiling and structure-based design to develop a selective, in vivo-compatible chemical probe of the DCLK1 kinase domain, DCLK1-IN-1. We demonstrate activity of DCLK1-IN-1 against clinically relevant patient-derived PDAC organoid models and use a combination of RNA-sequencing, proteomics and phosphoproteomics analysis to reveal that DCLK1 inhibition modulates proteins and pathways associated with cell motility in this context. DCLK1-IN-1 will serve as a versatile tool to investigate DCLK1 biology and establish its role in cancer.
Collapse
Affiliation(s)
- Fleur M Ferguson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Behnam Nabet
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Srivatsan Raghavan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yan Liu
- Departments of Biochemistry and Radiation Oncology, the University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alan L Leggett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Miljan Kuljanin
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Radha L Kalekar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Annan Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shuning He
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Raymond W S Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rita Sulahian
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lianbo Li
- Departments of Biochemistry and Radiation Oncology, the University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Emily J Poulin
- Cancer Research Institute and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ling Huang
- Cancer Research Institute and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jost Koren
- Department of Molecular and Human Genetics, Therapeutic Innovation Center Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nora Dieguez-Martinez
- Departament de Bioquímica i Biologia Molecular & Institut de Neurociencies, Facultat de Medicina. Universitat Autonoma de Barcelona, Bellaterra, Spain
| | - Sergio Espinosa
- Departament de Bioquímica i Biologia Molecular & Institut de Neurociencies, Facultat de Medicina. Universitat Autonoma de Barcelona, Bellaterra, Spain
| | | | | | | | - Ryoma Ohi
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Taebo Sim
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea and KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Nam Doo Kim
- NDBio Therapeutics Inc, Incheon, Republic of Korea
| | - Wayne Harshbarger
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- GSK Vaccines, Rockville, MD, USA
| | - Jose M Lizcano
- Departament de Bioquímica i Biologia Molecular & Institut de Neurociencies, Facultat de Medicina. Universitat Autonoma de Barcelona, Bellaterra, Spain
| | | | - Senthil Muthaswamy
- Cancer Research Institute and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Departments of Medicine and Pathology, Harvard Medical School, Boston, MA, USA
| | - Charles Y Lin
- Department of Molecular and Human Genetics, Therapeutic Innovation Center Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - A Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Kevin M Haigis
- Cancer Research Institute and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Digestive Disease Center, Harvard Medical School, Boston, MA, USA
| | - Joseph D Mancias
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew J Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kenneth D Westover
- Departments of Biochemistry and Radiation Oncology, the University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Li Z, Zhang X, Jin T, Hao J. Nicotine promotes activation of human pancreatic stellate cells through inducing autophagy via α7nAChR-mediated JAK2/STAT3 signaling pathway. Life Sci 2020; 243:117301. [PMID: 31953160 DOI: 10.1016/j.lfs.2020.117301] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/03/2020] [Accepted: 01/12/2020] [Indexed: 12/11/2022]
Abstract
AIM Pancreatic stellate cells (PSCs) are the main functional cells leading to pancreatic fibrosis. Nicotine is widely considered as an independent risk factor of pancreatic fibrosis, but the mechanism is still unclear. Our study was aimed to explore the effects of nicotine on human pancreatic stellate cells (hPSCs) and involved pathways. MATERIALS AND METHODS Primary human PSCs were cultured and treated with nicotine (0.1 μM and 1 μM). The proliferation, apoptosis, α-SMA expression, extracellular matrix metabolism and autophagy of hPSCs were detected by CCK-8 assay, flow cytometry, real-time PCR and Western blotting analysis. The α7nAChR-mediated JAK2/STAT3 signaling pathway was also examined, and an α7nAChR antagonist α-bungarotoxin (α-BTX) was used to perform inhibition experiments. KEY FINDINGS The proliferation, α-SMA expression and autophagy of hPSCs were significantly promoted by 1 μM nicotine. Meanwhile, the apoptosis of hPSCs was significantly reduced. The extracellular matrix metabolism of hPSCs was also regulated by nicotine. Moreover, the α7nAChR-mediated JAK2/STAT3 signaling pathway was activated by nicotine, this pathway and effects of nicotine can be blocked by α-BTX. SIGNIFICANCE Our finding suggests that nicotine can promote activation of human pancreatic stellate cells (hPSCs) through inducing autophagy via α7nAChR-mediated JAK2/STAT3 signaling pathway, providing a new insight into the mechanisms by which nicotine affects pancreatic fibrosis.
Collapse
Affiliation(s)
- Zhiren Li
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaoyun Zhang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Tong Jin
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jianyu Hao
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
25
|
Multiplexed proteome profiling of carbon source perturbations in two yeast species with SL-SP3-TMT. J Proteomics 2019; 210:103531. [PMID: 31626996 DOI: 10.1016/j.jprot.2019.103531] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/05/2019] [Accepted: 09/17/2019] [Indexed: 02/06/2023]
Abstract
Saccharomyces cerevisiae and Schizosaccharomyces pombe are the most commonly studied yeast model systems, yet comparisons of global proteome remodeling between these yeast species are scarce. Here, we profile the proteomes of S. cerevisiae and S. pombe cultured with either glucose or pyruvate as the sole carbon source to define common and distinctive alterations in the protein landscape across species. In addition, we develop an updated streamlined-tandem mass tag (SL-TMT) strategy that substitutes chemical-based precipitation with more versatile bead-based protein aggregation method (SP3) prior to enzymatic digestion and TMT labeling. Our new workflow, SL-SP3-TMT, allow for near-complete proteome profiles in a single experiment for each species. The data reveal expected alterations in protein abundance and differences between species, highlighted complete canonical biochemical pathways, and provided insight into previously uncharacterized proteins. The techniques used herein, namely SL-SP3-TMT, can be applied to virtually any experiment aiming to study remodeling of the proteome using a high-throughput, comprehensive, yet streamlined mass spectrometry-based strategy. SIGNIFICANCE: Saccharomyces cerevisiae and Schizosaccharomyces pombe are single-celled eukaryotes that diverged from a common ancestor over a period of 100 million years, such that evolution has driven fundamental differences between the two species. Cellular metabolism and the regulation thereof are vital for living organisms. Here, we hypothesize that large scale proteomic alterations are prevalent upon the substitution of glucose with another carbon source, in this case pyruvate. To efficiently process our samples, we developed an updated streamlined-tandem mass tag (SL-TMT) strategy with more versatile bead-based protein aggregation. The data revealed expected alterations in protein abundance and illustrated differences between species. We highlighted complete canonical biochemical pathways and provided insight into previously uncharacterized proteins.
Collapse
|
26
|
Azad F, Nejati V, Shalizar-Jalali A, Najafi G, Rahmani F. Antioxidant and anti-apoptotic effects of royal jelly against nicotine-induced testicular injury in mice. ENVIRONMENTAL TOXICOLOGY 2019; 34:708-718. [PMID: 30896085 DOI: 10.1002/tox.22737] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/28/2019] [Accepted: 02/07/2019] [Indexed: 06/09/2023]
Abstract
This study describes the effects of royal jelly (RJ) on testicular injury induced by nicotine (NIC) in mice. Thirty-six male BALB/c mice were randomly divided into six groups (n = 6). Group 1 received normal saline, group 2 received 100 mg/kgBW/day RJ, groups 3 and 4 received NIC at doses of 0.50 and 1.00 mg/kgBW/day, respectively, and groups 5 and 6 received NIC at doses of 0.50 and 1.00 mg/kg BW/day, respectively, plus RJ. Following 35 days, the serum level of testosterone, histopathological changes, germ cell apoptosis, proliferating cell nuclear antigen (PCNA), malondialdehyde (MDA) content, and antioxidant indexes including total antioxidant capacity (TAC) and catalase (CAT) activity were determined. In addition, the mitochondria-dependent apoptosis was investigated by assessing the Bcl-2, p53, and Caspase-3 mRNA levels expression by reverse transcription-PCR (RT-PCR). Compared to NIC receiving groups, the concomitant administration of RJ could protect the testosterone reduction and histological damages. After RJ treatment, the level of tissue MDA content decreased, while tissue TAC and CAT levels were remarkably increased compared to NIC-exposed groups. Remarkable higher TUNEL-positive germ cells and low PCNA index were observed in NIC receiving groups. Besides, the expression level of Bcl-2 was significantly higher and the p53 and Caspase-3 levels were significantly lower in the RJ co-administration groups than NIC-only receiving groups. Our results confirmed that RJ effectively protects the testis against NIC evoked damages by antioxidant and anti-apoptotic effects involving the up regulation of the antioxidant status, mitochondria-dependent apoptosis pathway prevention, and the proliferating activity improvement.
Collapse
Affiliation(s)
- Farnam Azad
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | - Vahid Nejati
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | - Ali Shalizar-Jalali
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Gholamreza Najafi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Fatemeh Rahmani
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| |
Collapse
|
27
|
Abed M, Verschueren E, Budayeva H, Liu P, Kirkpatrick DS, Reja R, Kummerfeld SK, Webster JD, Gierke S, Reichelt M, Anderson KR, Newman RJ, Roose-Girma M, Modrusan Z, Pektas H, Maltepe E, Newton K, Dixit VM. The Gag protein PEG10 binds to RNA and regulates trophoblast stem cell lineage specification. PLoS One 2019; 14:e0214110. [PMID: 30951545 PMCID: PMC6450627 DOI: 10.1371/journal.pone.0214110] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 03/15/2019] [Indexed: 01/03/2023] Open
Abstract
Peg10 (paternally expressed gene 10) is an imprinted gene that is essential for placental development. It is thought to derive from a Ty3-gyspy LTR (long terminal repeat) retrotransposon and retains Gag and Pol-like domains. Here we show that the Gag domain of PEG10 can promote vesicle budding similar to the HIV p24 Gag protein. Expressed in a subset of mouse endocrine organs in addition to the placenta, PEG10 was identified as a substrate of the deubiquitinating enzyme USP9X. Consistent with PEG10 having a critical role in placental development, PEG10-deficient trophoblast stem cells (TSCs) exhibited impaired differentiation into placental lineages. PEG10 expressed in wild-type, differentiating TSCs was bound to many cellular RNAs including Hbegf (Heparin-binding EGF-like growth factor), which is known to play an important role in placentation. Expression of Hbegf was reduced in PEG10-deficient TSCs suggesting that PEG10 might bind to and stabilize RNAs that are critical for normal placental development.
Collapse
Affiliation(s)
- Mona Abed
- Physiological Chemistry Department, Genentech, South San Francisco, California, United States of America
| | - Erik Verschueren
- Protein Chemistry Department, Genentech, South San Francisco, California, United States of America
| | - Hanna Budayeva
- Protein Chemistry Department, Genentech, South San Francisco, California, United States of America
| | - Peter Liu
- Protein Chemistry Department, Genentech, South San Francisco, California, United States of America
| | - Donald S. Kirkpatrick
- Protein Chemistry Department, Genentech, South San Francisco, California, United States of America
| | - Rohit Reja
- Bioinformatics and Computational Biology Department, Genentech, South San Francisco, California, United States of America
| | - Sarah K. Kummerfeld
- Bioinformatics and Computational Biology Department, Genentech, South San Francisco, California, United States of America
| | - Joshua D. Webster
- Pathology Department, Genentech, South San Francisco, California, United States of America
| | - Sarah Gierke
- Pathology Department, Genentech, South San Francisco, California, United States of America
| | - Mike Reichelt
- Pathology Department, Genentech, South San Francisco, California, United States of America
| | - Keith R. Anderson
- Molecular Biology Department, Genentech, South San Francisco, California, United States of America
| | - Robert J. Newman
- Molecular Biology Department, Genentech, South San Francisco, California, United States of America
| | - Merone Roose-Girma
- Molecular Biology Department, Genentech, South San Francisco, California, United States of America
| | - Zora Modrusan
- Molecular Biology Department, Genentech, South San Francisco, California, United States of America
| | - Hazal Pektas
- The Center for Reproductive Sciences, Division of Neonatology, University of California, San Francisco, California, United States of America
| | - Emin Maltepe
- The Center for Reproductive Sciences, Division of Neonatology, University of California, San Francisco, California, United States of America
| | - Kim Newton
- Physiological Chemistry Department, Genentech, South San Francisco, California, United States of America
| | - Vishva M. Dixit
- Physiological Chemistry Department, Genentech, South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
28
|
Krieger JR, Wybenga-Groot LE, Tong J, Bache N, Tsao MS, Moran MF. Evosep One Enables Robust Deep Proteome Coverage Using Tandem Mass Tags while Significantly Reducing Instrument Time. J Proteome Res 2019; 18:2346-2353. [DOI: 10.1021/acs.jproteome.9b00082] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
| | | | | | | | - Ming S. Tsao
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, Canada M5G 2C1
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King’s College Circle, Toronto, ON, Canada M5S 1A8
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON, Canada M5G 1L7
| | - Michael F. Moran
- Department of Molecular Genetics, University of Toronto, 1 King’s College Circle, Toronto, ON, Canada M5S 1A8
| |
Collapse
|
29
|
Muntel J, Kirkpatrick J, Bruderer R, Huang T, Vitek O, Ori A, Reiter L. Comparison of Protein Quantification in a Complex Background by DIA and TMT Workflows with Fixed Instrument Time. J Proteome Res 2019; 18:1340-1351. [DOI: 10.1021/acs.jproteome.8b00898] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jan Muntel
- Biognosys AG, Wagistrasse 21, 8952 Schlieren, Switzerland
| | - Joanna Kirkpatrick
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | | | - Ting Huang
- Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Olga Vitek
- Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Alessandro Ori
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Lukas Reiter
- Biognosys AG, Wagistrasse 21, 8952 Schlieren, Switzerland
| |
Collapse
|
30
|
Hu H, Zhao X, Ma J, Shangguan Y, Pan Z, Chen L, Zhang X, Wang H. Prenatal nicotine exposure retards osteoclastogenesis and endochondral ossification in fetal long bones in rats. Toxicol Lett 2018; 295:249-255. [PMID: 29981921 DOI: 10.1016/j.toxlet.2018.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 06/06/2018] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
Abstract
This study investigated the mechanisms underlying the retarded development of long bone in fetus by prenatal nicotine exposure (PNE) which had been demonstrated by our previous work. Nicotine (2.0 mg/kg.d) or saline was injected subcutaneously into pregnant rats every morning from gestational day (GD) 9 to 20. Fetal femurs or tibias were harvested for analysis on GD 20. We found massive accumulation of hypertrophic chondrocytes and a delayed formation of primary ossification center (POC) in the fetal femur or tibia of rat fetus after PNE, which was accompanied by a decreased amount of osteoclasts in the POC and up-regulated expression of osteoprotegerin (OPG) but by no obvious change in the expression of receptor activator of NF-κB ligand (RANKL). In primary osteoblastic cells, both nicotine (0, 162, 1620, 16,200 ng/ml) and corticosterone (0, 50, 250, 1250 nM) promoted the mRNA expression of OPG but concentration-dependently suppressed that of RANKL. Furthermore, blocking α4β2-nicotinic acetylcholine receptor (α4β2-nAChR) or glucocorticoid receptor rescued the above effects of nicotine and corticosterone, respectively. In conclusion, retarded osteoclastogenesis may contribute to delayed endochondral ossification in long bone in fetal rats with PNE. The adverse effects of PNE may be mediated via the direct effect of nicotine and indirect effect of maternal corticosterone on osteoblastic cells.
Collapse
Affiliation(s)
- Hang Hu
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Physiology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Xin Zhao
- Department of Physiology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Jing Ma
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Physiology, Basic Medical School of Wuhan University, Wuhan 430071, China
| | - Yangfan Shangguan
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhengqi Pan
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China; Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Liaobin Chen
- Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xianrong Zhang
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, China.
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
31
|
Navarrete-Perea J, Yu Q, Gygi SP, Paulo JA. Streamlined Tandem Mass Tag (SL-TMT) Protocol: An Efficient Strategy for Quantitative (Phospho)proteome Profiling Using Tandem Mass Tag-Synchronous Precursor Selection-MS3. J Proteome Res 2018; 17:2226-2236. [PMID: 29734811 DOI: 10.1021/acs.jproteome.8b00217] [Citation(s) in RCA: 254] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mass spectrometry (MS) coupled toisobaric labeling has developed rapidly into a powerful strategy for high-throughput protein quantification. Sample multiplexing and exceptional sensitivity allow for the quantification of tens of thousands of peptides and, by inference, thousands of proteins from multiple samples in a single MS experiment. Accurate quantification demands a consistent and robust sample-preparation strategy. Here, we present a detailed workflow for SPS-MS3-based quantitative abundance profiling of tandem mass tag (TMT)-labeled proteins and phosphopeptides that we have named the streamlined (SL)-TMT protocol. We describe a universally applicable strategy that requires minimal individual sample processing and permits the seamless addition of a phosphopeptide enrichment step ("mini-phos") with little deviation from the deep proteome analysis. To showcase our workflow, we profile the proteome of wild-type Saccharomyces cerevisiae yeast grown with either glucose or pyruvate as the carbon source. Here, we have established a streamlined TMT protocol that enables deep proteome and medium-scale phosphoproteome analysis.
Collapse
Affiliation(s)
- José Navarrete-Perea
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Qing Yu
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Steven P Gygi
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Joao A Paulo
- Department of Cell Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| |
Collapse
|
32
|
Paulo JA, Jedrychowski MP, Chouchani ET, Kazak L, Gygi SP. Multiplexed Isobaric Tag-Based Profiling of Seven Murine Tissues Following In Vivo Nicotine Treatment Using a Minimalistic Proteomics Strategy. Proteomics 2018; 18:e1700326. [PMID: 29660237 PMCID: PMC5992107 DOI: 10.1002/pmic.201700326] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/24/2017] [Indexed: 12/18/2022]
Abstract
Nicotine is a major addictive compound in tobacco and a component of smoking-related products, such as e-cigarettes. Once internalized, nicotine can perturb many cellular pathways and can induce alterations in proteins across different cell types; however, the mechanisms thereof remain undetermined. The authors hypothesize that both tissue-specific and global protein abundance alterations result from nicotine exposure. Presented here is the first proteomic profiling of multiple tissues from mice treated orally with nicotine. Proteins extracted from seven tissues (brain, heart, kidney, liver, lung, pancreas, and spleen) from treated (n = 5) and untreated control (n = 5) mice are assembled into a TMT10-plex experiment. A minimalistic proteomics strategy is employed using TMT reagents efficiently and centrifugation-based reversed-phase columns to streamline sample preparation. Combined, over 11 000 non-redundant proteins from over 138 000 different peptides are quantified in seven TMT10-plex experiments. Between 7 and 126 proteins are significantly altered in tissues from nicotine-exposed mice, 11 which are altered in two or more tissues. Our data showcase the vast extent of nicotine exposure across murine tissue.
Collapse
Affiliation(s)
- Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| | - Mark P. Jedrychowski
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, United States
| | - Edward T. Chouchani
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, United States
| | - Lawrence Kazak
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, United States
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| |
Collapse
|
33
|
Stepanova E, Gygi SP, Paulo JA. Filter-Based Protein Digestion (FPD): A Detergent-Free and Scaffold-Based Strategy for TMT Workflows. J Proteome Res 2018; 17:1227-1234. [PMID: 29402085 PMCID: PMC5984590 DOI: 10.1021/acs.jproteome.7b00840] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
High-throughput proteome profiling requires thorough optimization to achieve comprehensive analysis. We developed a filter aided sample preparation (FASP)-like, detergent-free method, termed Filter-Based Protein Digestion (FPD). We compared FPD to protein extraction methods commonly used in isobaric tag-based proteome profiling, namely trichloroacetic acid (TCA) and chloroform-methanol (C-M) precipitation. We divided a mammalian whole cell lysate from the SH-SY5Y neuroblastoma cell line for parallel protein processing with TCA (n = 3), C-M (n = 2), and FPD using either 10 kDa (n = 3) or 30 kDa (n = 3) molecular weight cutoff membranes. We labeled each sample with tandem mass tag (TMT) reagents to construct a TMT11-plex experiment. In total, 8654 proteins were quantified across all samples. Pairwise comparisons showed very little deviation for individual protein abundance measurements between the two FPD methods, whereas TCA and FPD showed the most difference. Specifically, membrane proteins were more readily quantified when samples were processed using TCA precipitation than other methods tested. However, globally, only 4% of proteins differed greater than 4-fold in the most divergent pair of protein extraction methods (i.e., FPD10 and TCA). We conclude that the detergent-free FPD strategy, particularly using the faster-flowing 30 kDa filter, is a seamless alteration to high-throughput TMT workflows.
Collapse
Affiliation(s)
- Ekaterina Stepanova
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| |
Collapse
|
34
|
Dumdie JN, Cho K, Ramaiah M, Skarbrevik D, Mora-Castilla S, Stumpo DJ, Lykke-Andersen J, Laurent LC, Blackshear PJ, Wilkinson MF, Cook-Andersen H. Chromatin Modification and Global Transcriptional Silencing in the Oocyte Mediated by the mRNA Decay Activator ZFP36L2. Dev Cell 2018; 44:392-402.e7. [PMID: 29408237 DOI: 10.1016/j.devcel.2018.01.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 06/21/2017] [Accepted: 01/05/2018] [Indexed: 12/19/2022]
Abstract
Global transcriptional silencing is a highly conserved mechanism central to the oocyte-to-embryo transition. We report the unexpected discovery that global transcriptional silencing in oocytes depends on an mRNA decay activator. Oocyte-specific loss of ZFP36L2 an RNA-binding protein that promotes AU-rich element-dependent mRNA decay prevents global transcriptional silencing and causes oocyte maturation and fertilization defects, as well as complete female infertility in the mouse. Single-cell RNA sequencing revealed that ZFP36L2 downregulates mRNAs encoding transcription and chromatin modification regulators, including a large group of mRNAs for histone demethylases targeting H3K4 and H3K9, which we show are bound and degraded by ZFP36L2. Oocytes lacking Zfp36l2 fail to accumulate histone methylation at H3K4 and H3K9, marks associated with the transcriptionally silent, developmentally competent oocyte state. Our results uncover a ZFP36L2-dependent mRNA decay mechanism that acts as a developmental switch during oocyte growth, triggering wide-spread shifts in chromatin modification and global transcription.
Collapse
Affiliation(s)
- Jennifer N Dumdie
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kyucheol Cho
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Madhuvanthi Ramaiah
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - David Skarbrevik
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sergio Mora-Castilla
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Deborah J Stumpo
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Jens Lykke-Andersen
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Louise C Laurent
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; Departments of Medicine and Biochemistry, Duke University Medical Center, Durham, NC 27703, USA
| | - Miles F Wilkinson
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Heidi Cook-Andersen
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
35
|
Zhao X, Huffman KE, Fujimoto J, Canales JR, Girard L, Nie G, Heymach JV, Wistuba II, Minna JD, Yu Y. Quantitative Proteomic Analysis of Optimal Cutting Temperature (OCT) Embedded Core-Needle Biopsy of Lung Cancer. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2017; 28:2078-2089. [PMID: 28752479 PMCID: PMC5693617 DOI: 10.1007/s13361-017-1706-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/03/2017] [Accepted: 05/04/2017] [Indexed: 06/07/2023]
Abstract
With recent advances in understanding the genomic underpinnings and oncogenic drivers of pathogenesis in different subtypes, it is increasingly clear that proper pretreatment diagnostics are essential for the choice of appropriate treatment options for non-small cell lung cancer (NSCLC). Tumor tissue preservation in optimal cutting temperature (OCT) compound is commonly used in the surgical suite. However, proteins recovered from OCT-embedded specimens pose a challenge for LC-MS/MS experiments, due to the large amounts of polymers present in OCT. Here we present a simple workflow for whole proteome analysis of OCT-embedded NSCLC tissue samples, which involves a simple trichloroacetic acid precipitation step. Comparisons of protein recovery between frozen versus OCT-embedded tissue showed excellent consistency with more than 9200 proteins identified. Using an isobaric labeling strategy, we quantified more than 5400 proteins in tumor versus normal OCT-embedded core needle biopsy samples. Gene ontology analysis indicated that a number of proliferative as well as squamous cell carcinoma (SqCC) marker proteins were overexpressed in the tumor, consistent with the patient's pathology based diagnosis of "poorly differentiated SqCC". Among the most downregulated proteins in the tumor sample, we noted a number of proteins with potential immunomodulatory functions. Finally, interrogation of the aberrantly expressed proteins using a candidate approach and cross-referencing with publicly available databases led to the identification of potential druggable targets in DNA replication and DNA damage repair pathways. We conclude that our approach allows LC-MS/MS proteomic analyses on OCT-embedded lung cancer specimens, opening the way to bring powerful proteomics into the clinic. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Xiaozheng Zhao
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kenneth E Huffman
- Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, Pharmacology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jamie Rodriguez Canales
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luc Girard
- Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, Pharmacology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guangjun Nie
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, People's Republic of China.
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| | - John V Heymach
- Department of Head and Neck and Thoracic Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Igacio I Wistuba
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, Pharmacology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yonghao Yu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
36
|
Whiteley AM, Prado MA, Peng I, Abbas AR, Haley B, Paulo JA, Reichelt M, Katakam A, Sagolla M, Modrusan Z, Lee DY, Roose-Girma M, Kirkpatrick DS, McKenzie BS, Gygi SP, Finley D, Brown EJ. Ubiquilin1 promotes antigen-receptor mediated proliferation by eliminating mislocalized mitochondrial proteins. eLife 2017; 6. [PMID: 28933694 PMCID: PMC5608509 DOI: 10.7554/elife.26435] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 08/22/2017] [Indexed: 12/14/2022] Open
Abstract
Ubiquilins (Ubqlns) are a family of ubiquitin receptors that promote the delivery of hydrophobic and aggregated ubiquitinated proteins to the proteasome for degradation. We carried out a proteomic analysis of a B cell lymphoma-derived cell line, BJAB, that requires UBQLN1 for survival to identify UBQLN1 client proteins. When UBQLN1 expression was acutely inhibited, 120 mitochondrial proteins were enriched in the cytoplasm, suggesting that the accumulation of mitochondrial client proteins in the absence of UBQLN1 is cytostatic. Using a Ubqln1−/− mouse strain, we found that B cell receptor (BCR) ligation of Ubqln1−/− B cells led to a defect in cell cycle entry. As in BJAB cells, mitochondrial proteins accumulated in BCR-stimulated cells, leading to protein synthesis inhibition and cell cycle block. Thus, UBQLN1 plays an important role in clearing mislocalized mitochondrial proteins upon cell stimulation, and its absence leads to suppression of protein synthesis and cell cycle arrest.
Collapse
Affiliation(s)
- Alexandra M Whiteley
- Department of Infectious Disease, Genentech, South San Francisco, United States.,Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Miguel A Prado
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Ivan Peng
- Department of Translational Immunology, Genentech, South San Francisco, United States
| | - Alexander R Abbas
- Department of Bioinformatics, Genentech, South San Francisco, United States
| | - Benjamin Haley
- Department of Molecular Biology, Genentech, South San Francisco, United States
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Mike Reichelt
- Department of Pathology, Genentech, South San Francisco, United States
| | - Anand Katakam
- Department of Pathology, Genentech, South San Francisco, United States
| | - Meredith Sagolla
- Department of Pathology, Genentech, South San Francisco, United States
| | - Zora Modrusan
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, South San Francisco, United States
| | - Dong Yun Lee
- Department of Infectious Disease, Genentech, South San Francisco, United States
| | - Merone Roose-Girma
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, South San Francisco, United States
| | - Donald S Kirkpatrick
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, South San Francisco, United States
| | - Brent S McKenzie
- Department of Translational Immunology, Genentech, South San Francisco, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Eric J Brown
- Department of Infectious Disease, Genentech, South San Francisco, United States
| |
Collapse
|
37
|
Sancilio S, Gallorini M, Cataldi A, Sancillo L, Rana RA, di Giacomo V. Modifications in Human Oral Fibroblast Ultrastructure, Collagen Production, and Lysosomal Compartment in Response to Electronic Cigarette Fluids. J Periodontol 2017; 88:673-680. [DOI: 10.1902/jop.2017.160629] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Silvia Sancilio
- Department of Pharmacy, University G. d’Annunzio, Chieti-Pescara, Italy
| | | | - Amelia Cataldi
- Department of Pharmacy, University G. d’Annunzio, Chieti-Pescara, Italy
| | - Laura Sancillo
- Department of Medicine and Aging Sciences, University G. d’Annunzio
| | - Rosa Alba Rana
- Department of Medicine and Aging Sciences, University G. d’Annunzio
| | | |
Collapse
|