105 results on '"Altelaar M"'
Search Results
2. Mesenchymal stromal/stem cells promote intestinal epithelium regeneration after chemotherapy-induced damage
- Author
-
Yetkin-Arik, B, Jansen, S A, Varderidou-Minasian, S, Westendorp, B, Skarp, K-P, Altelaar, M, Lindemans, C A, Lorenowicz, M J, Yetkin-Arik, B, Jansen, S A, Varderidou-Minasian, S, Westendorp, B, Skarp, K-P, Altelaar, M, Lindemans, C A, and Lorenowicz, M J
- Abstract
BACKGROUND: Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment for leukemia and a range of non-malignant disorders. The success of the therapy is hampered by occurrence of acute graft-versus-host disease (aGvHD); an inflammatory response damaging recipient organs, with gut, liver, and skin being the most susceptible. Intestinal GvHD injury is often a life-threatening complication in patients unresponsive to steroid treatment. Allogeneic mesenchymal stromal/stem cell (MSC) infusions are a promising potential treatment for steroid-resistant aGvHD. Data from our institution and others demonstrate rescue of approximately 40-50% of aGvHD patients with MSCs in Phase I, II studies and minor side effects. Although promising, better understanding of MSC mode of action and patient response to MSC-based therapy is essential to improve this lifesaving treatment.METHODS: Single cell human small intestine organoids were embedded in Matrigel, grown for 5 days and treated with busulfan for 48 h. Organoids damaged by treatment with busulfan or control organoids were co-cultured with 5000, 10,000, and 50,000 MSCs for 24 h, 48 h or 7 days and the analyses such as surface area determination, proliferation and apoptosis assessment, RNA sequencing and proteomics were performed.RESULTS: Here, we developed a 3D co-culture model of human small intestinal organoids and MSCs, which allows to study the regenerative effects of MSCs on intestinal epithelium in a more physiologically relevant setting than existing in vitro systems. Using this model we mimicked chemotherapy-mediated damage of the intestinal epithelium. The treatment with busulfan, the chemotherapeutic commonly used as conditioning regiment before the HSCT, affected pathways regulating epithelial to mesenchymal transition, proliferation, and apoptosis in small intestinal organoids, as shown by transcriptomic and proteomic analysis. The co-culture of busulfan-treated intestinal organoi
- Published
- 2024
3. Mesenchymal stromal/stem cells promote intestinal epithelium regeneration after chemotherapy-induced damage
- Author
-
MS Algemene Pediatrie Onderzoek 2, MS Neonatologie, SCT patientenzorg, Child Health, Infection & Immunity, Regenerative Medicine and Stem Cells, Cancer, CMM Groep Coffer, CMM, Yetkin-Arik, B., Jansen, S. A., Varderidou-Minasian, S., Westendorp, B., Skarp, K. P., Altelaar, M., Lindemans, C. A., Lorenowicz, M. J., MS Algemene Pediatrie Onderzoek 2, MS Neonatologie, SCT patientenzorg, Child Health, Infection & Immunity, Regenerative Medicine and Stem Cells, Cancer, CMM Groep Coffer, CMM, Yetkin-Arik, B., Jansen, S. A., Varderidou-Minasian, S., Westendorp, B., Skarp, K. P., Altelaar, M., Lindemans, C. A., and Lorenowicz, M. J.
- Published
- 2024
4. Mesenchymal stromal/stem cells promote intestinal epithelium regeneration after chemotherapy-induced damage
- Author
-
Pathobiologie, Cell Biology, Metabolism and Cancer, Afd Biomol.Mass Spect. and Proteomics, Cell Biology, Metabolism & Cancer - Cancer, Yetkin-Arik, B, Jansen, S A, Varderidou-Minasian, S, Westendorp, B, Skarp, K-P, Altelaar, M, Lindemans, C A, Lorenowicz, M J, Pathobiologie, Cell Biology, Metabolism and Cancer, Afd Biomol.Mass Spect. and Proteomics, Cell Biology, Metabolism & Cancer - Cancer, Yetkin-Arik, B, Jansen, S A, Varderidou-Minasian, S, Westendorp, B, Skarp, K-P, Altelaar, M, Lindemans, C A, and Lorenowicz, M J
- Published
- 2024
5. Identification of a tumor-specific allo-HLA–restricted γδTCR
- Author
-
Kierkels, G.J.J., Scheper, W., Meringa, A.D., Johanna, I., Beringer, D.X., Janssen, A., Schiffler, M., Aarts-Riemens, T., Kramer, L., Straetemans, T., Heijhuurs, S., Leusen, J.H.W., San José, E., Fuchs, K., Griffioen, M., Falkenburg, J.H., Bongiovanni, L., de Bruin, A., Vargas-Diaz, D., Altelaar, M., Heck, A.J.R., Shultz, L.D., Ishikawa, F., Nishimura, M.I., Sebestyén, Z., and Kuball, J.
- Published
- 2019
- Full Text
- View/download PDF
6. A comprehensive enhancer screen identifies TRAM2 as a key and novel mediator of YAP oncogenesis
- Author
-
Li, L. (Li), Ugalde, A.P. (Alejandro), Scheele, C.L.G.J. (Colinda L. G. J.), Dieter, S.M. (Sebastian M.), Nagel, C.R. (Remco), Ma, J. (Jin), Pataskar, A. (Abhijeet), Korkmaz, G. (Gozde), Elkon, R. (Ran), Chien, M.-P. (Miao-Ping), You, L. (Li), Su, P.-R. (Pin-Rui), Bleijerveld, O.B. (Onno), Altelaar, M. (Maarten), Momchev, L. (Lyubomir), Manber, Z. (Zohar), Han, R. (Ruiqi), Van Breugel, P.C. (Pieter C.), Lopes, R.F.M. (Rui), Dijke, P. (Peter) ten, van Rheenen, J. (Jacco), Agami, R. (Reuven), Li, L. (Li), Ugalde, A.P. (Alejandro), Scheele, C.L.G.J. (Colinda L. G. J.), Dieter, S.M. (Sebastian M.), Nagel, C.R. (Remco), Ma, J. (Jin), Pataskar, A. (Abhijeet), Korkmaz, G. (Gozde), Elkon, R. (Ran), Chien, M.-P. (Miao-Ping), You, L. (Li), Su, P.-R. (Pin-Rui), Bleijerveld, O.B. (Onno), Altelaar, M. (Maarten), Momchev, L. (Lyubomir), Manber, Z. (Zohar), Han, R. (Ruiqi), Van Breugel, P.C. (Pieter C.), Lopes, R.F.M. (Rui), Dijke, P. (Peter) ten, van Rheenen, J. (Jacco), and Agami, R. (Reuven)
- Abstract
Background: Frequent activation of the co-transcriptional factor YAP is observed in a large number of solid tumors. Activated YAP associates with enhancer loci via TEAD4-DNA-binding protein and stimulates cancer aggressiveness. Although thousands of YAP/TEAD4 binding-sites are annotated, their functional importance is unknown. Here, we aim at further identification of enhancer elements that are required for YAP functions. Results: We first apply genome-wide ChIP profiling of YAP to systematically identify enhancers that are bound by YAP/TEAD4. Next, we implement a genetic approach to uncover functions of YAP/TEAD4-associated enhancers, demonstrate its robustness, and use it to reveal a network of enhancers required for YAP-mediated proliferation. We focus on EnhancerTRAM2, as its target gene TRAM2 shows the strongest expression-correlation with YAP activity in nearly all tumor types. Interestingly, TRAM2 phenocopie
- Published
- 2021
- Full Text
- View/download PDF
7. A comprehensive enhancer screen identifies TRAM2 as a key and novel mediator of YAP oncogenesis
- Author
-
Li, L, Ugalde, AP, Scheele, C, Dieter, SM, van der Nagel, R, Ma, J, Pataskar, A, Korkmaz, G, Elkon, R, Chien, Miao, You, Li, Su, Pin-Rui, Bleijerveld, OB (Onno), Altelaar, M, Momchev, L, Manber, Z, Han, R Q, van Breugel, P C, Lopes, R (Rui Filipe Marques), ten Dijke, P, van Rheenen, J, Agami, Reuven, Li, L, Ugalde, AP, Scheele, C, Dieter, SM, van der Nagel, R, Ma, J, Pataskar, A, Korkmaz, G, Elkon, R, Chien, Miao, You, Li, Su, Pin-Rui, Bleijerveld, OB (Onno), Altelaar, M, Momchev, L, Manber, Z, Han, R Q, van Breugel, P C, Lopes, R (Rui Filipe Marques), ten Dijke, P, van Rheenen, J, and Agami, Reuven
- Abstract
Background: Frequent activation of the co-transcriptional factor YAP is observed in a large number of solid tumors. Activated YAP associates with enhancer loci via TEAD4-DNA-binding protein and stimulates cancer aggressiveness. Although thousands of YAP/TEAD4 binding-sites are annotated, their functional importance is unknown. Here, we aim at further identification of enhancer elements that are required for YAP functions. Results: We first apply genome-wide ChIP profiling of YAP to systematically identify enhancers that are bound by YAP/TEAD4. Next, we implement a genetic approach to uncover functions of YAP/TEAD4-associated enhancers, demonstrate its robustness, and use it to reveal a network of enhancers required for YAP-mediated proliferation. We focus on EnhancerTRAM2, as its target gene TRAM2 shows the strongest expression-correlation with YAP activity in nearly all tumor types. Interestingly, TRAM2 phenocopies the YAP-induced cell proliferation, migration, and invasion phenotypes and correlates with poor patient survival. Mechanistically, we identify FSTL-1 as a major direct client of TRAM2 that is involved in these phenotypes. Thus, TRAM2 is a key novel mediator of YAP-induced oncogenic proliferation and cellular invasiveness. Conclusions: YAP is a transcription co-factor that binds to thousands of enhancer loci and stimulates tumor aggressiveness. Using unbiased functional approaches, we dissect YAP enhancer network and characterize TRAM2 as a novel mediator of cellular proliferation, migration, and invasion. Our findings elucidate how YAP induces cancer aggressiveness and may assist diagnosis of cancer metastasis.
- Published
- 2021
8. Identification of a tumor-specific allo-HLA-restricted gamma delta TCR
- Author
-
Kierkels, G.J.J., Scheper, W., Meringa, A.D., Johanna, I., Beringer, D.X., Janssen, A., Schiffler, M., Aarts-Riemens, T., Kramer, L., Straetemans, T., Heijhuurs, S., Leusen, J.H.W., San Jose, E., Fuchs, K., Griffioen, M., Falkenburg, J.H., Bongiovanni, L., Bruin, A. de, Vargas-Diaz, D., Altelaar, M., Heck, A.J.R., Shultz, L.D., Ishikawa, F., Nishimura, M.I., Sebestyen, Z., and Kuball, J.
- Abstract
gamma delta T cells are key players in cancer immune surveillance because of their ability to recognize malignant transformed cells, which makes them promising therapeutic tools in the treatment of cancer. However, the biological mechanisms of how gamma delta T-cell receptors (TCRs) interact with their ligands are poorly understood. Within this context, we describe the novel allo-HLA-restricted and CD8 alpha-dependent V gamma 5V delta 1TCR. In contrast to the previous assumption of the general allo-HLA reactivity of a minor fraction of gamma delta TCRs, we show that classic anti-HLA-directed, gamma delta TCR-mediated reactivity can selectively act on hematological and solid tumor cells, while not harming healthy tissues in vitro and in vivo. We identified the molecular interface with proximity to the peptide-binding groove of HLA-A*24:02 as the essential determinant for recognition and describe the critical role of CD8 as a coreceptor. We conclude that alloreactive gamma delta T-cell repertoires provide therapeutic opportunities, either within the context of haplotransplantation or as individual gamma delta TCRs for genetic engineering of tumor-reactive T cells.
- Published
- 2019
9. Identification of a tumor-specific allo-HLA-restricted γδTCR
- Author
-
Kierkels, G J J, Scheper, W, Meringa, A D, Johanna, I, Beringer, D X, Janssen, A, Schiffler, M, Aarts-Riemens, T, Kramer, L, Straetemans, T, Heijhuurs, S, Leusen, J H W, San José, E, Fuchs, K, Griffioen, M, Falkenburg, J H, Bongiovanni, L, de Bruin, A, Vargas-Diaz, D, Altelaar, M, Heck, A J R, Shultz, L D, Ishikawa, F, Nishimura, M I, Sebestyén, Z, Kuball, J, Kierkels, G J J, Scheper, W, Meringa, A D, Johanna, I, Beringer, D X, Janssen, A, Schiffler, M, Aarts-Riemens, T, Kramer, L, Straetemans, T, Heijhuurs, S, Leusen, J H W, San José, E, Fuchs, K, Griffioen, M, Falkenburg, J H, Bongiovanni, L, de Bruin, A, Vargas-Diaz, D, Altelaar, M, Heck, A J R, Shultz, L D, Ishikawa, F, Nishimura, M I, Sebestyén, Z, and Kuball, J
- Published
- 2019
10. P.1.c.001 Interaction of the autism gene product contactin-6 with synaptic proteins
- Author
-
Zuko, A., primary, Oguro-Ando, A., additional, Kleijer, E., additional, Van der Zwaag, B., additional, Post, H., additional, Altelaar, M., additional, Heck, R., additional, Pasterkamp, R.J., additional, and Burbach, J.P.H., additional
- Published
- 2014
- Full Text
- View/download PDF
11. NAC regulates metabolism and cell fate in intestinal stem cells.
- Author
-
Ramalho S, Alkan F, Prekovic S, Jastrzebski K, Barberà EP, Hoekman L, Altelaar M, de Heus C, Liv N, Rodríguez-Colman MJ, Yilmaz M, van der Kammen R, Fedry J, de Gooijer MC, Suijkerbuijk SJE, Faller WJ, and Silva J
- Subjects
- Animals, Mice, Cell Differentiation, Organoids metabolism, Organoids cytology, Oxidative Phosphorylation, Intestinal Mucosa metabolism, Intestinal Mucosa cytology, Mitochondrial Proteins metabolism, Mitochondrial Proteins genetics, Stem Cells metabolism, Stem Cells cytology, Mitochondria metabolism, Ribosomes metabolism, Intestines cytology
- Abstract
Intestinal stem cells (ISCs) face the challenge of integrating metabolic demands with unique regenerative functions. Studies have shown an intricate interplay between metabolism and stem cell capacity; however, it is still not understood how this process is regulated. Combining ribosome profiling and CRISPR screening in intestinal organoids, we identify the nascent polypeptide-associated complex (NAC) as a key mediator of this process. Our findings suggest that NAC is responsible for relocalizing ribosomes to the mitochondria and regulating ISC metabolism. Upon NAC inhibition, intestinal cells show decreased import of mitochondrial proteins, which are needed for oxidative phosphorylation, and, consequently, enable the cell to maintain a stem cell identity. Furthermore, we show that overexpression of NACα is sufficient to drive mitochondrial respiration and promote ISC identity. Ultimately, our results reveal the pivotal role of NAC in regulating ribosome localization, mitochondrial metabolism, and ISC function, providing insights into the potential mechanism behind it.
- Published
- 2025
- Full Text
- View/download PDF
12. P-stalk ribosomes act as master regulators of cytokine-mediated processes.
- Author
-
Dopler A, Alkan F, Malka Y, van der Kammen R, Hoefakker K, Taranto D, Kocabay N, Mimpen I, Ramirez C, Malzer E, Isaeva OI, Kerkhoff M, Gangaev A, Silva J, Ramalho S, Hoekman L, Altelaar M, Beijersbergen R, Akkari L, Yewdell JW, Kvistborg P, and Faller WJ
- Subjects
- Animals, Humans, Mice, Phosphorylation, Transforming Growth Factor beta metabolism, RNA, Messenger metabolism, RNA, Messenger genetics, Mice, Inbred C57BL, Signal Transduction, Ribosomes metabolism, Cytokines metabolism, Protein Biosynthesis, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes immunology
- Abstract
Inflammatory cytokines are pivotal to immune responses. Upon cytokine exposure, cells enter an "alert state" that enhances their visibility to the immune system. Here, we identified an alert-state subpopulation of ribosomes defined by the presence of the P-stalk. We show that P-stalk ribosomes (PSRs) are formed in response to cytokines linked to tumor immunity, and this is at least partially mediated by P-stalk phosphorylation. PSRs are involved in the preferential translation of mRNAs vital for the cytokine response via the more efficient translation of transmembrane domains of receptor molecules involved in cytokine-mediated processes. Importantly, loss of the PSR inhibits CD8+ T cell recognition and killing, and inhibitory cytokines like transforming growth factor β (TGF-β) hinder PSR formation, suggesting that the PSR is a central regulatory hub upon which multiple signals converge. Thus, the PSR is an essential mediator of the cellular rewiring that occurs following cytokine exposure via the translational regulation of this process., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. TMED inhibition suppresses cell surface PD-1 expression and overcomes T cell dysfunction.
- Author
-
Vredevoogd DW, Apriamashvili G, Levy PL, Sinha S, Huinen ZR, Visser NL, de Bruijn B, Boshuizen J, van Hal-van Veen SE, Ligtenberg MA, Bleijerveld OB, Lin CP, Díaz-Gómez J, Sánchez SD, Markovits E, Simon Nieto J, van Vliet A, Krijgsman O, Markel G, Besser MJ, Altelaar M, Ruppin E, and Peeper DS
- Subjects
- Animals, Mice, Humans, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes drug effects, Tumor Microenvironment, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor metabolism
- Abstract
Background: Blockade of the programmed cell death protein 1 (PD-1) immune checkpoint (ICB) is revolutionizing cancer therapy, but little is known about the mechanisms governing its expression on CD8 T cells. Because PD-1 is induced during activation of T cells, we set out to uncover regulators whose inhibition suppresses PD-1 abundance without adversely impacting on T cell activation., Methods: To identify PD-1 regulators in an unbiased fashion, we performed a whole-genome, fluorescence-activated cell sorting (FACS)-based CRISPR-Cas9 screen in primary murine CD8 T cells. A dual-readout design using the activation marker CD137 allowed us to uncouple genes involved in PD-1 regulation from those governing general T cell activation., Results: We found that the inactivation of one of several members of the TMED/EMP24/GP25L/p24 family of transport proteins, most prominently TMED10, reduced PD-1 cell surface abundance, thereby augmenting T cell activity. Another client protein was cytotoxic T lymphocyte-associated protein 4 (CTLA-4), which was also suppressed by TMED inactivation. Treatment with TMED inhibitor AGN192403 led to lysosomal degradation of the TMED-PD-1 complex and reduced PD-1 abundance in tumor-infiltrating CD8 T cells (TIL) in mice, thus reversing T cell dysfunction. Clinically corroborating these findings, single-cell RNA analyses revealed a positive correlation between TMED expression in CD8 TIL, and both a T cell dysfunction signature and lack of ICB response. Similarly, patients receiving a TIL product with high TMED expression had a shorter overall survival., Conclusion: Our results uncover a novel mechanism of PD-1 regulation, and identify a pharmacologically tractable target whose inhibition suppresses PD-1 abundance and T cell dysfunction., Competing Interests: Competing interests: DSP and MAL are co-founders, shareholders and advisors of Flindr Tx (previously Immagene), which is unrelated to this study. The other authors declare no competing interests., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
14. Axonal endoplasmic reticulum tubules control local translation via P180/RRBP1-mediated ribosome interactions.
- Author
-
Koppers M, Özkan N, Nguyen HH, Jurriens D, McCaughey J, Nguyen DTM, Li CH, Stucchi R, Altelaar M, MacGillavry HD, Kapitein LC, Hoogenraad CC, and Farías GG
- Subjects
- Animals, Humans, Rats, Cells, Cultured, Neurons metabolism, Ribosomal Proteins metabolism, Ribosomal Proteins genetics, Carrier Proteins genetics, Carrier Proteins metabolism, Axons metabolism, Endoplasmic Reticulum metabolism, Hippocampus metabolism, Protein Biosynthesis, Ribosomes metabolism, RNA, Messenger genetics, RNA, Messenger metabolism
- Abstract
Local mRNA translation in axons is critical for the spatiotemporal regulation of the axonal proteome. A wide variety of mRNAs are localized and translated in axons; however, how protein synthesis is regulated at specific subcellular sites in axons remains unclear. Here, we establish that the axonal endoplasmic reticulum (ER) supports axonal translation in developing rat hippocampal cultured neurons. Axonal ER tubule disruption impairs local translation and ribosome distribution. Using nanoscale resolution imaging, we find that ribosomes make frequent contacts with axonal ER tubules in a translation-dependent manner and are influenced by specific extrinsic cues. We identify P180/RRBP1 as an axonally distributed ribosome receptor that regulates local translation and binds to mRNAs enriched for axonal membrane proteins. Importantly, the impairment of axonal ER-ribosome interactions causes defects in axon morphology. Our results establish a role for the axonal ER in dynamically localizing mRNA translation, which is important for proper neuron development., Competing Interests: Declaration of interests C.C.H. is an employee of Genentech, a member of the Roche group., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
15. Region-specific and age-related differences in astrocytes in the human brain.
- Author
-
Man JHK, Breur M, van Gelder CAGH, Marcon G, Maderna E, Giaccone G, Altelaar M, van der Knaap MS, and Bugiani M
- Subjects
- Humans, Adult, Aged, Young Adult, Middle Aged, Aged, 80 and over, Child, Infant, Child, Preschool, Adolescent, Infant, Newborn, Brain cytology, Brain pathology, Brain metabolism, Proteomics, Male, Female, Cell Count, Astrocytes pathology, Astrocytes metabolism, Aging pathology, Aging physiology, Gray Matter pathology, Gray Matter cytology, White Matter pathology, White Matter cytology
- Abstract
Astrocyte heterogeneity and its relation to aging in the normal human brain remain poorly understood. We here analyzed astrocytes in gray and white matter brain tissues obtained from donors ranging in age between the neonatal period to over 100 years. We show that astrocytes are differently distributed with higher density in the white matter. This regional difference in cellular density becomes less prominent with age. Additionally, we confirm the presence of morphologically distinct astrocytes, with gray matter astrocytes being morphologically more complex. Notably, gray matter astrocytes morphologically change with age, while white matter astrocytes remain relatively consistent in morphology. Using regional mass spectrometry-based proteomics, we did, however, identify astrocyte specific proteins with regional differences in abundance, reflecting variation in cellular density or expression level. Importantly, the expression of some astrocyte specific proteins region-dependently decreases with age. Taken together, we provide insights into region- and age-related differences in astrocytes in the human brain., Competing Interests: Declaration of Competing Interest The authors declare that they have no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. Dual role of proliferating cell nuclear antigen monoubiquitination in facilitating Fanconi anemia-mediated interstrand crosslink repair.
- Author
-
Shah R, Aslam MA, Spanjaard A, de Groot D, Zürcher LM, Altelaar M, Hoekman L, Pritchard CEJ, Pilzecker B, van den Berk PCM, and Jacobs H
- Abstract
The Fanconi anemia (FA) repair pathway governs repair of highly genotoxic DNA interstrand crosslinks (ICLs) and relies on translesion synthesis (TLS). TLS is facilitated by REV1 or site-specific monoubiquitination of proliferating cell nuclear antigen (PCNA) (PCNA-Ub) at lysine 164 (K164). A Pcna
K164R/K164R but not Rev1-/- mutation renders mammals hypersensitive to ICLs. Besides the FA pathway, alternative pathways have been associated with ICL repair (1, 2), though the decision making between those remains elusive. To study the dependence and relevance of PCNA-Ub in FA repair, we intercrossed PcnaK164R/+ ; Fancg-/+ mice. A combined mutation ( PcnaK164R/K164R ; Fancg-/- ) was found embryonically lethal. RNA-seq of primary double-mutant (DM) mouse embryonic fibroblasts (MEFs) revealed elevated levels of replication stress-induced checkpoints. To exclude stress-induced confounders, we utilized a Trp53 knock-down to obtain a model to study ICL repair in depth. Regarding ICL-induced cell toxicity, cell cycle arrest, and replication fork progression, single-mutant and DM MEFs were found equally sensitive, establishing PCNA-Ub to be critical for FA-ICL repair. Immunoprecipitation and spectrometry-based analysis revealed an unknown role of PCNA-Ub in excluding mismatch recognition complex MSH2/MSH6 from being recruited to ICLs. In conclusion, our results uncovered a dual function of PCNA-Ub in ICL repair, i.e. exclude MSH2/MSH6 recruitment to channel the ICL toward canonical FA repair, in addition to its established role in coordinating TLS opposite the unhooked ICL., (© The Author(s) 2024. Published by Oxford University Press on behalf of National Academy of Sciences.)- Published
- 2024
- Full Text
- View/download PDF
17. Proteomic dissection of vanishing white matter pathogenesis.
- Author
-
Man JHK, Zarekiani P, Mosen P, de Kok M, Debets DO, Breur M, Altelaar M, van der Knaap MS, and Bugiani M
- Subjects
- Animals, Mice, Humans, Corpus Callosum metabolism, Corpus Callosum pathology, Eukaryotic Initiation Factor-2B metabolism, Eukaryotic Initiation Factor-2B genetics, Brain metabolism, Brain pathology, Mice, Inbred C57BL, Cerebellum metabolism, Cerebellum pathology, Proteomics, Proteome metabolism, Leukoencephalopathies metabolism, Leukoencephalopathies genetics, Leukoencephalopathies pathology, Disease Models, Animal, White Matter metabolism, White Matter pathology
- Abstract
Vanishing white matter (VWM) is a leukodystrophy caused by biallelic pathogenic variants in eukaryotic translation initiation factor 2B. To date, it remains unclear which factors contribute to VWM pathogenesis. Here, we investigated the basis of VWM pathogenesis using the 2b5
ho mouse model. We first mapped the temporal proteome in the cerebellum, corpus callosum, cortex, and brainstem of 2b5ho and wild-type (WT) mice. Protein changes observed in 2b5ho mice were then cross-referenced with published proteomic datasets from VWM patient brain tissue to define alterations relevant to the human disease. By comparing 2b5ho mice with their region- and age-matched WT counterparts, we showed that the proteome in the cerebellum and cortex of 2b5ho mice was already dysregulated prior to pathology development, whereas proteome changes in the corpus callosum only occurred after pathology onset. Remarkably, protein changes in the brainstem were transient, indicating that a compensatory mechanism might occur in this region. Importantly, 2b5ho mouse brain proteome changes reflect features well-known in VWM. Comparison of the 2b5ho mouse and VWM patient brain proteomes revealed shared changes. These could represent changes that contribute to the disease or even drive its progression in patients. Taken together, we show that the 2b5ho mouse brain proteome is affected in a region- and time-dependent manner. We found that the 2b5ho mouse model partly replicates the human disease at the protein level, providing a resource to study aspects of VWM pathogenesis by highlighting alterations from early to late disease stages, and those that possibly drive disease progression., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
18. Predicting treatment outcome using kinome activity profiling in HER2+ breast cancer biopsies.
- Author
-
Debets DO, de Graaf EL, Liefaard MC, Sonke GS, Lips EH, Ressa A, and Altelaar M
- Abstract
In this study, we measured the kinase activity profiles of 32 pre-treatment tumor biopsies of HER2-positive breast cancer patients. The aim of this study was to assess the prognostic potential of kinase activity levels, to identify potential mechanisms of resistance and to predict treatment success of HER2-targeted therapy combined with chemotherapy. Indeed, our system-wide kinase activity analysis allowed us to link kinase activity to treatment response. Overall, high kinase activity in the HER2-pathway was associated with good treatment outcome. We found eleven kinases differentially regulated between treatment outcome groups, including well-known players in therapy resistance, such as p38a, ERK, and FAK, and an unreported one, namely MARK1. Lastly, we defined an optimal signature of four kinases in a multiple logistic regression diagnostic test for prediction of treatment outcome (AUC = 0.926). This kinase signature showed high sensitivity and specificity, indicating its potential as predictive biomarker for treatment success of HER2-targeted therapy., Competing Interests: M.A. is inventor of a patent named Method for monitoring kinase activity in a sample, patent number: 11624082., (© 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
19. Multimodal stimulation screens reveal unique and shared genes limiting T cell fitness.
- Author
-
Lin CP, Levy PL, Alflen A, Apriamashvili G, Ligtenberg MA, Vredevoogd DW, Bleijerveld OB, Alkan F, Malka Y, Hoekman L, Markovits E, George A, Traets JJH, Krijgsman O, van Vliet A, Poźniak J, Pulido-Vicuña CA, de Bruijn B, van Hal-van Veen SE, Boshuizen J, van der Helm PW, Díaz-Gómez J, Warda H, Behrens LM, Mardesic P, Dehni B, Visser NL, Marine JC, Markel G, Faller WJ, Altelaar M, Agami R, Besser MJ, and Peeper DS
- Subjects
- Humans, CD8-Positive T-Lymphocytes, CRISPR-Cas Systems, Neoplasms genetics
- Abstract
Genes limiting T cell antitumor activity may serve as therapeutic targets. It has not been systematically studied whether there are regulators that uniquely or broadly contribute to T cell fitness. We perform genome-scale CRISPR-Cas9 knockout screens in primary CD8 T cells to uncover genes negatively impacting fitness upon three modes of stimulation: (1) intense, triggering activation-induced cell death (AICD); (2) acute, triggering expansion; (3) chronic, causing dysfunction. Besides established regulators, we uncover genes controlling T cell fitness either specifically or commonly upon differential stimulation. Dap5 ablation, ranking highly in all three screens, increases translation while enhancing tumor killing. Loss of Icam1-mediated homotypic T cell clustering amplifies cell expansion and effector functions after both acute and intense stimulation. Lastly, Ctbp1 inactivation induces functional T cell persistence exclusively upon chronic stimulation. Our results functionally annotate fitness regulators based on their unique or shared contribution to traits limiting T cell antitumor activity., Competing Interests: Declaration of interests C.L., P.L., A.A., G.A., M.A.L., D.W.V., and D.S.P. are named as inventors on a patent filed by the Netherlands Cancer Institute and Oncode on the findings described in this manuscript. D.S.P. and M.A.L. are co-founders, shareholders, and advisors of Immagene., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
20. Innovative strategies for measuring kinase activity to accelerate the next wave of novel kinase inhibitors.
- Author
-
Veth TS, Kannegieter NM, de Graaf EL, Ruijtenbeek R, Joore J, Ressa A, and Altelaar M
- Subjects
- Humans, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Antineoplastic Agents pharmacology, Neoplasms drug therapy
- Abstract
The development of protein kinase inhibitors (PKIs) has gained significance owing to their therapeutic potential for diseases like cancer. In addition, there has been a rise in refining kinase activity assays, each possessing unique biological and analytical characteristics crucial for PKI development. However, the PKI development pipeline experiences high attrition rates and approved PKIs exhibit unexploited potential because of variable patient responses. Enhancing PKI development efficiency involves addressing challenges related to understanding the PKI mechanism of action and employing biomarkers for precision medicine. Selecting appropriate kinase activity assays for these challenges can overcome these attrition rate issues. This review delves into the current obstacles in kinase inhibitor development and elucidates kinase activity assays that can provide solutions., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
21. CAMSAPs and nucleation-promoting factors control microtubule release from γ-TuRC.
- Author
-
Rai D, Song Y, Hua S, Stecker K, Monster JL, Yin V, Stucchi R, Xu Y, Zhang Y, Chen F, Katrukha EA, Altelaar M, Heck AJR, Wieczorek M, Jiang K, and Akhmanova A
- Subjects
- Microtubules metabolism, Microtubule-Organizing Center metabolism, Cytoskeleton metabolism, Tubulin metabolism, Microtubule-Associated Proteins genetics, Microtubule-Associated Proteins metabolism
- Abstract
γ-Tubulin ring complex (γ-TuRC) is the major microtubule-nucleating factor. After nucleation, microtubules can be released from γ-TuRC and stabilized by other proteins, such as CAMSAPs, but the biochemical cross-talk between minus-end regulation pathways is poorly understood. Here we reconstituted this process in vitro using purified components. We found that all CAMSAPs could bind to the minus ends of γ-TuRC-attached microtubules. CAMSAP2 and CAMSAP3, which decorate and stabilize growing minus ends but not the minus-end tracking protein CAMSAP1, induced microtubule release from γ-TuRC. CDK5RAP2, a γ-TuRC-interactor, and CLASP2, a regulator of microtubule growth, strongly stimulated γ-TuRC-dependent microtubule nucleation, but only CDK5RAP2 suppressed CAMSAP binding to γ-TuRC-anchored minus ends and their release. CDK5RAP2 also improved selectivity of γ-tubulin-containing complexes for 13- rather than 14-protofilament microtubules in microtubule-capping assays. Knockout and overexpression experiments in cells showed that CDK5RAP2 inhibits the formation of CAMSAP2-bound microtubules detached from the microtubule-organizing centre. We conclude that CAMSAPs can release newly nucleated microtubules from γ-TuRC, whereas nucleation-promoting factors can differentially regulate this process., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
22. Phosphoprotein dynamics of interacting T cells and tumor cells by HySic.
- Author
-
Ibáñez-Molero S, Pruijs JTM, Atmopawiro A, Wang F, Terry AM, Altelaar M, Peeper DS, and Stecker KE
- Subjects
- Humans, Signal Transduction, Cell Communication, Phosphorylation, Isotope Labeling methods, Proteome metabolism, Phosphoproteins metabolism, Neoplasms
- Abstract
Functional interactions between cytotoxic T cells and tumor cells are central to anti-cancer immunity. However, our understanding of the proteins involved is limited. Here, we present HySic (hybrid quantification of stable isotope labeling by amino acids in cell culture [SILAC]-labeled interacting cells) as a method to quantify protein and phosphorylation dynamics between and within physically interacting cells. Using co-cultured T cells and tumor cells, we directly measure the proteome and phosphoproteome of engaged cells without the need for physical separation. We identify proteins whose abundance or activation status changes upon T cell:tumor cell interaction and validate our method with established signal transduction pathways including interferon γ (IFNγ) and tumor necrosis factor (TNF). Furthermore, we identify the RHO/RAC/PAK1 signaling pathway to be activated upon cell engagement and show that pharmacologic inhibition of PAK1 sensitizes tumor cells to T cell killing. Thus, HySic is a simple method to study rapid protein signaling dynamics in physically interacting cells that is easily extended to other biological systems., Competing Interests: Declaration of interests S.I.-M. and D.S.P. are named inventors on patent P097110NL, which is unrelated to this work. D.S.P. is co-founder, shareholder, and advisor of Immagene, which is unrelated to this work. M.A. is currently lead scientific officer for Amigon, which has no relation to this work., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
23. Proteomics on malignant pleural effusions reveals ERα loss in metastatic breast cancer associates with SGK1-NDRG1 deregulation.
- Author
-
Mayayo-Peralta I, Debets DO, Prekovic S, Schuurman K, Beerthuijzen S, Almekinders M, Sanders J, Moelans CB, Saleiro S, Wesseling J, van Diest PJ, Henrique R, Jerónimo C, Altelaar M, and Zwart W
- Subjects
- Female, Humans, Estrogen Receptor alpha metabolism, Glucocorticoids therapeutic use, Proteomics, Breast Neoplasms pathology, Pleural Effusion, Malignant
- Abstract
Breast cancer (BCa) is a highly heterogeneous disease, with hormone receptor status being a key factor in patient prognostication and treatment decision-making. The majority of primary tumours are positive for oestrogen receptor alpha (ERα), which plays a key role in tumorigenesis and disease progression, and represents the major target for treatment of BCa. However, around one-third of patients with ERα-positive BCa relapse and progress into the metastatic stage, with 20% of metastatic cases characterised by loss of ERα expression after endocrine treatment, known as ERα-conversion. It remains unclear whether ERα-converted cancers are biologically similar to bona fide ERα-negative disease and which signalling cascades compensate for ERα loss and drive tumour progression. To better understand the biological changes that occur in metastatic BCa upon ERα loss, we performed (phospho)proteomics analysis of 47 malignant pleural effusions derived from 37 BCa patients, comparing ERα-positive, ERα-converted and ERα-negative cases. Our data revealed that the loss of ERα-dependency in this metastatic site leads to only a partial switch to an ERα-negative molecular phenotype, with preservation of a luminal-like proteomic landscape. Furthermore, we found evidence for decreased activity of several key kinases, including serum/glucocorticoid regulated kinase 1 (SGK1), in ERα-converted metastases. Loss of SGK1 substrate phosphorylation may compensate for the loss of ERα-dependency in advanced disease and exposes a potential therapeutic vulnerability that may be exploited in treating these patients., (© 2023 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
24. Luminal breast cancer identity is determined by loss of glucocorticoid receptor activity.
- Author
-
Prekovic S, Chalkiadakis T, Roest M, Roden D, Lutz C, Schuurman K, Opdam M, Hoekman L, Abbott N, Tesselaar T, Wajahat M, Dwyer AR, Mayayo-Peralta I, Gomez G, Altelaar M, Beijersbergen R, Győrffy B, Young L, Linn S, Jonkers J, Tilley W, Hickey T, Vareslija D, Swarbrick A, and Zwart W
- Subjects
- Female, Humans, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Receptors, Estrogen genetics, Receptors, Estrogen metabolism, Receptors, Glucocorticoid genetics, Receptors, Glucocorticoid metabolism, Breast Neoplasms genetics, Breast Neoplasms drug therapy
- Abstract
Glucocorticoid receptor (GR) is a transcription factor that plays a crucial role in cancer biology. In this study, we utilized an in silico-designed GR activity signature to demonstrate that GR relates to the proliferative capacity of numerous primary cancer types. In breast cancer, the GR activity status determines luminal subtype identity and has implications for patient outcomes. We reveal that GR engages with estrogen receptor (ER), leading to redistribution of ER on the chromatin. Notably, GR activation leads to upregulation of the ZBTB16 gene, encoding for a transcriptional repressor, which controls growth in ER-positive breast cancer and associates with prognosis in luminal A patients. In relation to ZBTB16's repressive nature, GR activation leads to epigenetic remodeling and loss of histone acetylation at sites proximal to cancer-driving genes. Based on these findings, epigenetic inhibitors reduce viability of ER-positive breast cancer cells that display absence of GR activity. Our findings provide insights into how GR controls ER-positive breast cancer growth and may have implications for patients' prognostication and provide novel therapeutic candidates for breast cancer treatment., (© 2023 Netherlands Cancer Institute. Published under the terms of the CC BY 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
25. MYC is a clinically significant driver of mTOR inhibitor resistance in breast cancer.
- Author
-
Bhin J, Yemelyanenko J, Chao X, Klarenbeek S, Opdam M, Malka Y, Hoekman L, Kruger D, Bleijerveld O, Brambillasca CS, Sprengers J, Siteur B, Annunziato S, van Haren MJ, Martin NI, van de Ven M, Peters D, Agami R, Linn SC, Boven E, Altelaar M, Jonkers J, Zingg D, and Wessels LFA
- Subjects
- Humans, Animals, Mice, Female, MTOR Inhibitors, Phosphatidylinositol 3-Kinases, Proto-Oncogene Proteins c-akt, TOR Serine-Threonine Kinases, Breast Neoplasms drug therapy
- Abstract
Targeting the PI3K-AKT-mTOR pathway is a promising therapeutic strategy for breast cancer treatment. However, low response rates and development of resistance to PI3K-AKT-mTOR inhibitors remain major clinical challenges. Here, we show that MYC activation drives resistance to mTOR inhibitors (mTORi) in breast cancer. Multiomic profiling of mouse invasive lobular carcinoma (ILC) tumors revealed recurrent Myc amplifications in tumors that acquired resistance to the mTORi AZD8055. MYC activation was associated with biological processes linked to mTORi response and counteracted mTORi-induced translation inhibition by promoting translation of ribosomal proteins. In vitro and in vivo induction of MYC conferred mTORi resistance in mouse and human breast cancer models. Conversely, AZD8055-resistant ILC cells depended on MYC, as demonstrated by the synergistic effects of mTORi and MYCi combination treatment. Notably, MYC status was significantly associated with poor response to everolimus therapy in metastatic breast cancer patients. Thus, MYC is a clinically relevant driver of mTORi resistance that may stratify breast cancer patients for mTOR-targeted therapies., (© 2023 Bhin et al.)
- Published
- 2023
- Full Text
- View/download PDF
26. Deep (phospho)proteomics profiling of pre- treatment needle biopsies identifies signatures of treatment resistance in HER2 + breast cancer.
- Author
-
Debets DO, Stecker KE, Piskopou A, Liefaard MC, Wesseling J, Sonke GS, Lips EH, and Altelaar M
- Subjects
- Humans, Female, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Proteomics, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Neoadjuvant Therapy, Biopsy, Needle, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms metabolism
- Abstract
Patients with early-stage HER2-overexpressing breast cancer struggle with treatment resistance in 20%-40% of cases. More information is needed to predict HER2 therapy response and resistance in vivo. In this study, we perform (phospho)proteomics analysis of pre-treatment HER2
+ needle biopsies of early-stage invasive breast cancer to identify molecular signatures predictive of treatment response to trastuzumab, pertuzumab, and chemotherapy. Our data show that accurate quantification of the estrogen receptor (ER) and HER2 biomarkers, combined with the assessment of associated biological features, has the potential to enable better treatment outcome prediction. In addition, we identify cellular mechanisms that potentially precondition tumors to resist therapy. We find proteins with expression changes that correlate with resistance and constitute to a strong predictive signature for treatment success in our patient cohort. Our results highlight the multifactorial nature of drug resistance in vivo and demonstrate the necessity of deep tumor profiling., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
27. PAXIP1 and STAG2 converge to maintain 3D genome architecture and facilitate promoter/enhancer contacts to enable stress hormone-dependent transcription.
- Author
-
Mayayo-Peralta I, Gregoricchio S, Schuurman K, Yavuz S, Zaalberg A, Kojic A, Abbott N, Geverts B, Beerthuijzen S, Siefert J, Severson TM, van Baalen M, Hoekman L, Lieftink C, Altelaar M, Beijersbergen RL, Houtsmuller AB, Prekovic S, and Zwart W
- Abstract
How steroid hormone receptors (SHRs) regulate transcriptional activity remains partly understood. Upon activation, SHRs bind the genome together with a co-regulator repertoire, crucial to induce gene expression. However, it remains unknown which components of the SHR-recruited co-regulator complex are essential to drive transcription following hormonal stimuli. Through a FACS-based genome-wide CRISPR screen, we functionally dissected the Glucocorticoid Receptor (GR) complex. We describe a functional cross-talk between PAXIP1 and the cohesin subunit STAG2, critical for regulation of gene expression by GR. Without altering the GR cistrome, PAXIP1 and STAG2 depletion alter the GR transcriptome, by impairing the recruitment of 3D-genome organization proteins to the GR complex. Importantly, we demonstrate that PAXIP1 is required for stability of cohesin on chromatin, its localization to GR-occupied sites, and maintenance of enhancer-promoter interactions. In lung cancer, where GR acts as tumor suppressor, PAXIP1/STAG2 loss enhances GR-mediated tumor suppressor activity by modifying local chromatin interactions. All together, we introduce PAXIP1 and STAG2 as novel co-regulators of GR, required to maintain 3D-genome architecture and drive the GR transcriptional programme following hormonal stimuli., (© The Author(s) 2023. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2023
- Full Text
- View/download PDF
28. A cyclin-dependent kinase-mediated phosphorylation switch of disordered protein condensation.
- Author
-
Valverde JM, Dubra G, Phillips M, Haider A, Elena-Real C, Fournet A, Alghoul E, Chahar D, Andrés-Sanchez N, Paloni M, Bernadó P, van Mierlo G, Vermeulen M, van den Toorn H, Heck AJR, Constantinou A, Barducci A, Ghosh K, Sibille N, Knipscheer P, Krasinska L, Fisher D, and Altelaar M
- Subjects
- Phosphorylation, Cell Cycle, Cyclin-Dependent Kinase 2 metabolism, Cyclin-Dependent Kinases metabolism, Cell Cycle Proteins metabolism
- Abstract
Cell cycle transitions result from global changes in protein phosphorylation states triggered by cyclin-dependent kinases (CDKs). To understand how this complexity produces an ordered and rapid cellular reorganisation, we generated a high-resolution map of changing phosphosites throughout unperturbed early cell cycles in single Xenopus embryos, derived the emergent principles through systems biology analysis, and tested them by biophysical modelling and biochemical experiments. We found that most dynamic phosphosites share two key characteristics: they occur on highly disordered proteins that localise to membraneless organelles, and are CDK targets. Furthermore, CDK-mediated multisite phosphorylation can switch homotypic interactions of such proteins between favourable and inhibitory modes for biomolecular condensate formation. These results provide insight into the molecular mechanisms and kinetics of mitotic cellular reorganisation., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
29. Elucidating Fibroblast Growth Factor-Induced Kinome Dynamics Using Targeted Mass Spectrometry and Dynamic Modeling.
- Author
-
Veth TS, Francavilla C, Heck AJR, and Altelaar M
- Subjects
- Phosphorylation, Cell Proliferation, Mass Spectrometry, Fibroblast Growth Factors pharmacology, Fibroblast Growth Factor 2 pharmacology
- Abstract
Fibroblast growth factors (FGFs) are paracrine or endocrine signaling proteins that, activated by their ligands, elicit a wide range of health and disease-related processes, such as cell proliferation and the epithelial-to-mesenchymal transition. The detailed molecular pathway dynamics that coordinate these responses have remained to be determined. To elucidate these, we stimulated MCF-7 breast cancer cells with either FGF2, FGF3, FGF4, FGF10, or FGF19. Following activation of the receptor, we quantified the kinase activity dynamics of 44 kinases using a targeted mass spectrometry assay. Our system-wide kinase activity data, supplemented with (phospho)proteomics data, reveal ligand-dependent distinct pathway dynamics, elucidate the involvement of not earlier reported kinases such as MARK, and revise some of the pathway effects on biological outcomes. In addition, logic-based dynamic modeling of the kinome dynamics further verifies the biological goodness-of-fit of the predicted models and reveals BRAF-driven activation upon FGF2 treatment and ARAF-driven activation upon FGF4 treatment., Competing Interests: Conflicts of interest The authors declare that they have no conflict of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
30. Natural deletion of mouse carboxylesterases Ces1c/d/e impacts drug metabolism and metabolic syndrome development.
- Author
-
Gan C, Wang J, Wang Y, Martínez-Chávez A, Hillebrand M, de Vries N, Beukers J, Lebre MC, Wagenaar E, Rosing H, Klarenbeek S, Bleijerveld OB, Song JY, Altelaar M, Beijnen JH, and Schinkel AH
- Subjects
- Animals, Female, Male, Mice, Inflammation, Irinotecan, Lipids, Mammals, Obesity metabolism, Carboxylic Ester Hydrolases genetics, Carboxylic Ester Hydrolases metabolism, Metabolic Syndrome, Prodrugs
- Abstract
Mammalian carboxylesterase 1 enzymes can hydrolyze many xenobiotic chemicals and endogenous lipids. We here identified and characterized a mouse strain (FVB/NKI) in which three of the eight Ces1 genes were spontaneously deleted, removing Ces1c and Ces1e partly, and Ces1d entirely. We studied the impact of this Ces1c/d/e deficiency on drug and lipid metabolism and homeostasis. Ces1c/d/e
-/- mice showed strongly impaired conversion of the anticancer prodrug irinotecan to its active metabolite SN-38 in plasma, spleen and lung. Plasma hydrolysis of the oral anticancer prodrug capecitabine to 5-DFCR was also profoundly reduced in Ces1c/d/e-/- mice. Our findings resolved previously unexplained FVB/NKI pharmacokinetic anomalies. On a medium-fat diet, Ces1c/d/e-/- female mice exhibited moderately higher body weight, mild inflammation in gonadal white adipose tissue (gWAT), and increased lipid load in brown adipose tissue (BAT). Ces1c/d/e-/- males showed more pronounced inflammation in gWAT and an increased lipid load in BAT. On a 5-week high-fat diet exposure, Ces1c/d/e deficiency predisposed to developing obesity, enlarged and fatty liver, glucose intolerance and insulin resistance, with severe inflammation in gWAT and increased lipid load in BAT. Hepatic proteomics analysis revealed that the acute phase response, involved in the dynamic cycle of immunometabolism, was activated in these Ces1c/d/e-/- mice. This may contribute to the obesity-related chronic inflammation and adverse metabolic disease in this strain. While Ces1c/d/e deficiency clearly exacerbated metabolic syndrome development, long-term (18-week) high-fat diet exposure overwhelmed many, albeit not all, observed phenotypic differences., Competing Interests: Declaration of Competing Interest The authors declare that there are no conflicts of interest., (Copyright © 2023 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
31. Optimized Suspension Trapping Method for Phosphoproteomics Sample Preparation.
- Author
-
Wang F, Veth T, Kuipers M, Altelaar M, and Stecker KE
- Subjects
- Mass Spectrometry, Proteolysis, Proteins analysis, Proteomics methods
- Abstract
A successful mass spectrometry-based phosphoproteomics analysis relies on effective sample preparation strategies. Suspension trapping (S-Trap) is a novel, rapid, and universal method of sample preparation that is increasingly applied in bottom-up proteomics studies. However, the performance of the S-Trap protocol for phosphoproteomics studies is unclear. In the existing S-Trap protocol, the addition of phosphoric acid (PA) and methanol buffer creates a fine protein suspension to capture proteins on a filter and is a critical step for subsequent protein digestion. Herein, we demonstrate that this addition of PA is detrimental to downstream phosphopeptide enrichment, rendering the standard S-Trap protocol suboptimal for phosphoproteomics. In this study, the performance of the S-Trap digestion for proteomics and phosphoproteomics is systematically evaluated in large-scale and small-scale samples. The results of this comparative analysis show that an optimized S-Trap approach, where trifluoroacetic acid is substituted for PA, is a simple and effective method to prepare samples for phosphoproteomics. Our optimized S-Trap protocol is applied to extracellular vesicles to demonstrate superior sample preparation workflow for low-abundance, membrane-rich samples.
- Published
- 2023
- Full Text
- View/download PDF
32. Regional vulnerability of brain white matter in vanishing white matter.
- Author
-
Man JHK, van Gelder CAGH, Breur M, Molenaar D, Abbink T, Altelaar M, Bugiani M, and van der Knaap MS
- Subjects
- Child, Humans, Child, Preschool, Proteome metabolism, Brain pathology, Oxidative Phosphorylation, White Matter pathology, Leukoencephalopathies pathology
- Abstract
Vanishing white matter (VWM) is a leukodystrophy that primarily manifests in young children. In this disease, the brain white matter is differentially affected in a predictable pattern with telencephalic brain areas being most severely affected, while others remain allegedly completely spared. Using high-resolution mass spectrometry-based proteomics, we investigated the proteome patterns of the white matter in the severely affected frontal lobe and normal appearing pons in VWM and control cases to identify molecular bases underlying regional vulnerability. By comparing VWM patients to controls, we identified disease-specific proteome patterns. We showed substantial changes in both the VWM frontal and pons white matter at the protein level. Side-by-side comparison of brain region-specific proteome patterns further revealed regional differences. We found that different cell types were affected in the VWM frontal white matter than in the pons. Gene ontology and pathway analyses identified involvement of region specific biological processes, of which pathways involved in cellular respiratory metabolism were overarching features. In the VWM frontal white matter, proteins involved in glycolysis/gluconeogenesis and metabolism of various amino acids were decreased compared to controls. By contrast, in the VWM pons white matter, we found a decrease in proteins involved in oxidative phosphorylation. Taken together, our data show that brain regions are affected in parallel in VWM, but to different degrees. We found region-specific involvement of different cell types and discovered that cellular respiratory metabolism is likely to be differentially affected across white matter regions in VWM. These region-specific changes help explain regional vulnerability to pathology in VWM., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
33. A genome-wide CRISPR screen in human prostate cancer cells reveals drivers of macrophage-mediated cell killing and positions AR as a tumor-intrinsic immunomodulator.
- Author
-
Zaalberg A, Minnee E, Mayayo-Peralta I, Schuurman K, Gregoricchio S, van Schaik TA, Hoekman L, Li D, Corey E, Janssen H, Lieftink C, Prekovic S, Altelaar M, Nelson PS, Beijersbergen RL, Zwart W, and Bergman A
- Abstract
The crosstalk between prostate cancer (PCa) cells and the tumor microenvironment plays a pivotal role in disease progression and metastasis and could provide novel opportunities for patient treatment. Macrophages are the most abundant immune cells in the prostate tumor microenvironment (TME) and are capable of killing tumor cells. To identify genes in the tumor cells that are critical for macrophage-mediated killing, we performed a genome-wide co-culture CRISPR screen and identified AR, PRKCD, and multiple components of the NF-κB pathway as hits, whose expression in the tumor cell are essential for being targeted and killed by macrophages. These data position AR signaling as an immunomodulator, and confirmed by androgen-deprivation experiments, that rendered hormone-deprived tumor cells resistant to macrophage-mediated killing. Proteomic analyses showed a downregulation of oxidative phosphorylation in the PRKCD- and IKBKG-KO cells compared to the control, suggesting impaired mitochondrial function, which was confirmed by electron microscopy analyses. Furthermore, phosphoproteomic analyses revealed that all hits impaired ferroptosis signaling, which was validated transcriptionally using samples from a neoadjuvant clinical trial with the AR-inhibitor enzalutamide. Collectively, our data demonstrate that AR functions together with the PRKCD and the NF-κB pathway to evade macrophage-mediated killing. As hormonal intervention represents the mainstay therapy for treatment of prostate cancer patients, our findings may have direct implications and provide a plausible explanation for the clinically observed persistence of tumor cells despite androgen deprivation therapy.
- Published
- 2023
- Full Text
- View/download PDF
34. Ubiquitinome Profiling Reveals in Vivo UBE2D3 Targets and Implicates UBE2D3 in Protein Quality Control.
- Author
-
Yalçin Z, Koot D, Bezstarosti K, Salas-Lloret D, Bleijerveld OB, Boersma V, Falcone M, González-Prieto R, Altelaar M, Demmers JAA, and Jacobs JJL
- Subjects
- Ubiquitination, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitin-Protein Ligases metabolism, Proteome metabolism, Ubiquitin metabolism
- Abstract
Ubiquitination has crucial roles in many cellular processes, and dysregulation of ubiquitin machinery enzymes can result in various forms of pathogenesis. Cells only have a limited set of ubiquitin-conjugating (E2) enzymes to support the ubiquitination of many cellular targets. As individual E2 enzymes have many different substrates and interactions between E2 enzymes and their substrates can be transient, it is challenging to define all in vivo substrates of an individual E2 and the cellular processes it affects. Particularly challenging in this respect is UBE2D3, an E2 enzyme with promiscuous activity in vitro but less defined roles in vivo. Here, we set out to identify in vivo targets of UBE2D3 by using stable isotope labeling by amino acids in cell culture-based and label-free quantitative ubiquitin diGly proteomics to study global proteome and ubiquitinome changes associated with UBE2D3 depletion. UBE2D3 depletion changed the global proteome, with the levels of proteins from metabolic pathways, in particular retinol metabolism, being the most affected. However, the impact of UBE2D3 depletion on the ubiquitinome was much more prominent. Interestingly, molecular pathways related to mRNA translation were the most affected. Indeed, we find that ubiquitination of the ribosomal proteins RPS10 and RPS20, critical for ribosome-associated protein quality control, is dependent on UBE2D3. We show by Targets of Ubiquitin Ligases Identified by Proteomics 2 methodology that RPS10 and RPS20 are direct targets of UBE2D3 and demonstrate that the catalytic activity of UBE2D3 is required to ubiquitinate RPS10 in vivo. In addition, our data suggest that UBE2D3 acts at multiple levels in autophagic protein quality control. Collectively, our findings show that depletion of an E2 enzyme in combination with quantitative diGly-based ubiquitinome profiling is a powerful tool to identify new in vivo E2 substrates, as we have done here for UBE2D3. Our work provides an important resource for further studies on the in vivo functions of UBE2D3., Competing Interests: Conflict of interest The authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
35. Identification of Protein Complexes by Integrating Protein Abundance and Interaction Features Using a Deep Learning Strategy.
- Author
-
Li B, Altelaar M, and van Breukelen B
- Subjects
- Proteomics methods, Protein Subunits, Machine Learning, Mass Spectrometry, Deep Learning
- Abstract
Many essential cellular functions are carried out by multi-protein complexes that can be characterized by their protein-protein interactions. The interactions between protein subunits are critically dependent on the strengths of their interactions and their cellular abundances, both of which span orders of magnitude. Despite many efforts devoted to the global discovery of protein complexes by integrating large-scale protein abundance and interaction features, there is still room for improvement. Here, we integrated >7000 quantitative proteomic samples with three published affinity purification/co-fractionation mass spectrometry datasets into a deep learning framework to predict protein-protein interactions (PPIs), followed by the identification of protein complexes using a two-stage clustering strategy. Our deep-learning-technique-based classifier significantly outperformed recently published machine learning prediction models and in the process captured 5010 complexes containing over 9000 unique proteins. The vast majority of proteins in our predicted complexes exhibited low or no tissue specificity, which is an indication that the observed complexes tend to be ubiquitously expressed throughout all cell types and tissues. Interestingly, our combined approach increased the model sensitivity for low abundant proteins, which amongst other things allowed us to detect the interaction of MCM10, which connects to the replicative helicase complex via the MCM6 protein. The integration of protein abundances and their interaction features using a deep learning approach provided a comprehensive map of protein-protein interactions and a unique perspective on possible novel protein complexes.
- Published
- 2023
- Full Text
- View/download PDF
36. Systemic LRG1 Expression in Melanoma is Associated with Disease Progression and Recurrence.
- Author
-
Hoefsmit EP, Völlmy F, Rozeman EA, Reijers ILM, Versluis JM, Hoekman L, van Akkooi ACJ, Long GV, Schadendorf D, Dummer R, Altelaar M, and Blank CU
- Subjects
- Humans, Disease Progression, Prognosis, Biomarkers, Glycoproteins, Proteomics, Melanoma
- Abstract
The response rates upon neoadjuvant immune checkpoint blockade (ICB) in stage III melanoma are higher as compared with stage IV disease. Given that successful ICB depends on systemic immune response, we hypothesized that systemic immune suppression might be a mechanism responsible for lower response rates in late-stage disease, and also potentially with disease recurrence in early-stage disease. Plasma and serum samples of cohorts of patients with melanoma were analyzed for circulating proteins using mass spectrometry proteomic profiling and Olink proteomic assay. A cohort of paired samples of patients with stage III that progressed to stage IV disease ( n = 64) was used to identify markers associated with higher tumor burden. Baseline patient samples from the OpACIN-neo study ( n = 83) and PRADO study ( n = 49; NCT02977052) were used as two independent cohorts to analyze whether the potential identified markers are also associated with disease recurrence after neoadjuvant ICB therapy. When comparing baseline proteins overlapping between patients with progressive disease and patients with recurrent disease, we found leucine-rich alpha-2-glycoprotein 1 (LRG1) to be associated with worse prognosis. Especially nonresponder patients to neoadjuvant ICB (OpACIN-neo) with high LRG1 expression had a poor outcome with an estimated 36-month event-free survival of 14% as compared with 83% for nonresponders with a low LRG1 expression ( P = 0.014). This finding was validated in an independent cohort ( P = 0.0021). LRG1 can be used as a biomarker to identify patients with high risk for disease progression and recurrence, and might be a target to be combined with neoadjuvant ICB., Significance: LRG1 could serve as a potential target and as a biomarker to identify patients with high risk for disease recurrence, and consequently benefit from additional therapies and intensive follow-up., (© 2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
37. Cortical Pathology in Vanishing White Matter.
- Author
-
Man JHK, van Gelder CAGH, Breur M, Okkes D, Molenaar D, van der Sluis S, Abbink T, Altelaar M, van der Knaap MS, and Bugiani M
- Subjects
- Humans, Astrocytes metabolism, Proteomics, Mitochondria metabolism, White Matter pathology, Leukoencephalopathies genetics
- Abstract
Vanishing white matter (VWM) is classified as a leukodystrophy with astrocytes as primary drivers in its pathogenesis. Magnetic resonance imaging has documented the progressive thinning of cortices in long-surviving patients. Routine histopathological analyses, however, have not yet pointed to cortical involvement in VWM. Here, we provide a comprehensive analysis of the VWM cortex. We employed high-resolution-mass-spectrometry-based proteomics and immunohistochemistry to gain insight into possible molecular disease mechanisms in the cortices of VWM patients. The proteome analysis revealed 268 differentially expressed proteins in the VWM cortices compared to the controls. A majority of these proteins formed a major protein interaction network. A subsequent gene ontology analysis identified enrichment for terms such as cellular metabolism, particularly mitochondrial activity. Importantly, some of the proteins with the most prominent changes in expression were found in astrocytes, indicating cortical astrocytic involvement. Indeed, we confirmed that VWM cortical astrocytes exhibit morphological changes and are less complex in structure than control cells. Our findings also suggest that these astrocytes are immature and not reactive. Taken together, we provide insights into cortical involvement in VWM, which has to be taken into account when developing therapeutic strategies.
- Published
- 2022
- Full Text
- View/download PDF
38. Drug-Induced Epigenomic Plasticity Reprograms Circadian Rhythm Regulation to Drive Prostate Cancer toward Androgen Independence.
- Author
-
Linder S, Hoogstraat M, Stelloo S, Eickhoff N, Schuurman K, de Barros H, Alkemade M, Bekers EM, Severson TM, Sanders J, Huang CF, Morova T, Altintas UB, Hoekman L, Kim Y, Baca SC, Sjöström M, Zaalberg A, Hintzen DC, de Jong J, Kluin RJC, de Rink I, Giambartolomei C, Seo JH, Pasaniuc B, Altelaar M, Medema RH, Feng FY, Zoubeidi A, Freedman ML, Wessels LFA, Butler LM, Lack NA, van der Poel H, Bergman AM, and Zwart W
- Subjects
- ARNTL Transcription Factors genetics, Cell Line, Tumor, Circadian Rhythm, Drug Resistance, Neoplasm genetics, Epigenomics, Humans, Male, Nitriles therapeutic use, Receptors, Androgen genetics, Androgens pharmacology, Androgens therapeutic use, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology
- Abstract
In prostate cancer, androgen receptor (AR)-targeting agents are very effective in various disease stages. However, therapy resistance inevitably occurs, and little is known about how tumor cells adapt to bypass AR suppression. Here, we performed integrative multiomics analyses on tissues isolated before and after 3 months of AR-targeting enzalutamide monotherapy from patients with high-risk prostate cancer enrolled in a neoadjuvant clinical trial. Transcriptomic analyses demonstrated that AR inhibition drove tumors toward a neuroendocrine-like disease state. Additionally, epigenomic profiling revealed massive enzalutamide-induced reprogramming of pioneer factor FOXA1 from inactive chromatin sites toward active cis-regulatory elements that dictate prosurvival signals. Notably, treatment-induced FOXA1 sites were enriched for the circadian clock component ARNTL. Posttreatment ARNTL levels were associated with patients' clinical outcomes, and ARNTL knockout strongly decreased prostate cancer cell growth. Our data highlight a remarkable cistromic plasticity of FOXA1 following AR-targeted therapy and revealed an acquired dependency on the circadian regulator ARNTL, a novel candidate therapeutic target., Significance: Understanding how prostate cancers adapt to AR-targeted interventions is critical for identifying novel drug targets to improve the clinical management of treatment-resistant disease. Our study revealed an enzalutamide-induced epigenomic plasticity toward prosurvival signaling and uncovered the circadian regulator ARNTL as an acquired vulnerability after AR inhibition, presenting a novel lead for therapeutic development. See related commentary by Zhang et al., p. 2017. This article is highlighted in the In This Issue feature, p. 2007., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
39. The centriolar satellite protein Cfap53 facilitates formation of the zygotic microtubule organizing center in the zebrafish embryo.
- Author
-
Willekers S, Tessadori F, van der Vaart B, Henning HH, Stucchi R, Altelaar M, Roelen BAJ, Akhmanova A, and Bakkers J
- Subjects
- Animals, Centrosome metabolism, Male, Semen metabolism, Tubulin metabolism, Zebrafish metabolism, Centrioles metabolism, Microtubule-Organizing Center metabolism
- Abstract
In embryos of most animal species, the zygotic centrosome is assembled by the centriole derived from the sperm cell and pericentriolar proteins present in the oocyte. This zygotic centrosome acts as a microtubule organizing center (MTOC) to assemble the sperm aster and mitotic spindle. As MTOC formation has been studied mainly in adult cells, very little is known about the formation of the zygotic MTOC. Here, we show that zebrafish (Danio rerio) embryos lacking either maternal or paternal Cfap53, a centriolar satellite protein, arrest during the first cell cycle. Although Cfap53 is dispensable for sperm aster function, it aids proper formation of the mitotic spindle. During cell division, Cfap53 colocalizes with γ-tubulin and with other centrosomal and centriolar satellite proteins at the MTOC. Furthermore, we find that γ-tubulin localization at the MTOC is impaired in the absence of Cfap53. Based on these results, we propose a model in which Cfap53 deposited in the oocyte and the sperm participates in the organization of the zygotic MTOC to allow mitotic spindle formation., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
40. Ribosome impairment regulates intestinal stem cell identity via ZAKɑ activation.
- Author
-
Silva J, Alkan F, Ramalho S, Snieckute G, Prekovic S, Garcia AK, Hernández-Pérez S, van der Kammen R, Barnum D, Hoekman L, Altelaar M, Zwart W, Suijkerbuijk SJE, Bekker-Jensen S, and Faller WJ
- Subjects
- Animals, Intestine, Small metabolism, Intestines, Mice, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Ribosomes metabolism, Intestinal Mucosa metabolism, Stem Cells metabolism
- Abstract
The small intestine is a rapidly proliferating organ that is maintained by a small population of Lgr5-expressing intestinal stem cells (ISCs). However, several Lgr5-negative ISC populations have been identified, and this remarkable plasticity allows the intestine to rapidly respond to both the local environment and to damage. However, the mediators of such plasticity are still largely unknown. Using intestinal organoids and mouse models, we show that upon ribosome impairment (driven by Rptor deletion, amino acid starvation, or low dose cyclohexamide treatment) ISCs gain an Lgr5-negative, fetal-like identity. This is accompanied by a rewiring of metabolism. Our findings suggest that the ribosome can act as a sensor of nutrient availability, allowing ISCs to respond to the local nutrient environment. Mechanistically, we show that this phenotype requires the activation of ZAKɑ, which in turn activates YAP, via SRC. Together, our data reveals a central role for ribosome dynamics in intestinal stem cells, and identify the activation of ZAKɑ as a critical mediator of stem cell identity., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
41. Author Correction: Tryptophan depletion results in tryptophan-to-phenylalanine substitutants.
- Author
-
Pataskar A, Champagne J, Nagel R, Kenski J, Laos M, Michaux J, Pak HS, Bleijerveld OB, Mordente K, Navarro JM, Blommaert N, Nielsen MM, Lovecchio D, Stone E, Georgiou G, de Gooijer MC, van Tellingen O, Altelaar M, Joosten RP, Perrakis A, Olweus J, Bassani-Sternberg M, Peeper DS, and Agami R
- Published
- 2022
- Full Text
- View/download PDF
42. Differentiated kidney tubular cell-derived extracellular vesicles enhance maturation of tubuloids.
- Author
-
Lindoso RS, Yousef Yengej FA, Voellmy F, Altelaar M, Mancheño Juncosa E, Tsikari T, Ammerlaan CME, Van Balkom BWM, Rookmaaker MB, Verhaar MC, and Masereeuw R
- Subjects
- Epithelial Cells, Kidney metabolism, Kidney Tubules, Proximal metabolism, Extracellular Vesicles metabolism, Proteomics
- Abstract
The prevalence of end-stage kidney disease (ESKD) is rapidly increasing with the need for regenerative therapies. Adult stem cell derived kidney tubuloids have the potential to functionally mimic the adult kidney tubule, but still lack the expression of important transport proteins needed for waste removal. Here, we investigated the potential of extracellular vesicles (EVs) obtained from matured kidney tubular epithelial cells to modulate in vitro tubuloids functional maturation. We focused on organic anion transporter 1 (OAT1), one of the most important proteins involved in endogenous waste excretion. First, we show that EVs from engineered proximal tubule cells increased the expression of several transcription factors and epithelial transporters, resulting in improved OAT1 transport capacity. Next, a more in-depth proteomic data analysis showed that EVs can trigger various biological pathways, including mesenchymal-to-epithelial transition, which is crucial in the tubular epithelial maturation. Moreover, we demonstrated that the combination of EVs and tubuloid-derived cells can be used as part of a bioartificial kidney to generate a tight polarized epithelial monolayer with formation of dense cilia structures. In conclusion, EVs from kidney tubular epithelial cells can phenotypically improve in vitro tubuloid maturation, thereby enhancing their potential as functional units in regenerative or renal replacement therapies., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
43. The encephalomyocarditis virus Leader promotes the release of virions inside extracellular vesicles via the induction of secretory autophagy.
- Author
-
van der Grein SG, Defourny KAY, Rabouw HH, Goerdayal SS, van Herwijnen MJC, Wubbolts RW, Altelaar M, van Kuppeveld FJM, and Nolte-'t Hoen ENM
- Subjects
- Autophagy, Viral Proteins metabolism, Virion metabolism, Encephalomyocarditis virus metabolism, Extracellular Vesicles metabolism
- Abstract
Naked viruses can escape host cells before the induction of lysis via release in extracellular vesicles (EVs). These nanosized EVs cloak the secreted virus particles in a host-derived membrane, which alters virus-host interactions that affect infection efficiency and antiviral immunity. Currently, little is known about the viral and host factors regulating this form of virus release. Here, we assessed the role of the encephalomyocarditis virus (EMCV) Leader protein, a 'viral security protein' that subverts the host antiviral response. EV release upon infection with wildtype virus or a Leader-deficient mutant was characterized at the single particle level using high-resolution flow cytometry. Inactivation of the Leader abolished EV induction during infection and strongly reduced EV-enclosed virus release. We demonstrate that the Leader promotes the release of virions within EVs by stimulating a secretory arm of autophagy. This newly discovered role of the EMCV Leader adds to the variety of mechanisms via which this protein affects virus-host interactions. Moreover, these data provide first evidence for a crucial role of a non-structural viral protein in the non-lytic release of picornaviruses via packaging in EVs., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
44. RNF31 inhibition sensitizes tumors to bystander killing by innate and adaptive immune cells.
- Author
-
Zhang Z, Kong X, Ligtenberg MA, van Hal-van Veen SE, Visser NL, de Bruijn B, Stecker K, van der Helm PW, Kuilman T, Hoefsmit EP, Vredevoogd DW, Apriamashvili G, Baars B, Voest EE, Klarenbeek S, Altelaar M, and Peeper DS
- Subjects
- Killer Cells, Natural, Tumor Necrosis Factor-alpha metabolism, Ubiquitination, Tumor Escape, Ubiquitin-Protein Ligases genetics
- Abstract
Tumor escape mechanisms for immunotherapy include deficiencies in antigen presentation, diminishing adaptive CD8
+ T cell antitumor activity. Although innate natural killer (NK) cells are triggered by loss of MHC class I, their response is often inadequate. To increase tumor susceptibility to both innate and adaptive immune elimination, we performed parallel genome-wide CRISPR-Cas9 knockout screens under NK and CD8+ T cell pressure. We identify all components, RNF31, RBCK1, and SHARPIN, of the linear ubiquitination chain assembly complex (LUBAC). Genetic and pharmacologic ablation of RNF31, an E3 ubiquitin ligase, strongly sensitizes cancer cells to NK and CD8+ T cell killing. This occurs in a tumor necrosis factor (TNF)-dependent manner, causing loss of A20 and non-canonical IKK complexes from TNF receptor complex I. A small-molecule RNF31 inhibitor sensitizes colon carcinoma organoids to TNF and greatly enhances bystander killing of MHC antigen-deficient tumor cells. These results merit exploration of RNF31 inhibition as a clinical pharmacological opportunity for immunotherapy-refractory cancers., Competing Interests: Declaration of interests D.S.P. is co-founder, shareholder, and advisor of Immagene. M.A.L. is co-founder, shareholder, and CEO of Immagene. The other authors declare no competing interests., (Copyright © 2022 Netherlands Cancer Institute. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
45. Angiomotin isoform 2 promotes binding of PALS1 to KIF13B at primary cilia and regulates ciliary length and signaling.
- Author
-
Morthorst SK, Nielsen C, Farinelli P, Anvarian Z, Rasmussen CBR, Serra-Marques A, Grigoriev I, Altelaar M, Fürstenberg N, Ludwig A, Akhmanova A, Christensen ST, and Pedersen LB
- Subjects
- Cilia metabolism, Guanylate Kinases, Hedgehog Proteins metabolism, Protein Isoforms, Angiomotins, Membrane Proteins metabolism
- Abstract
The kinesin-3 motor KIF13B functions in endocytosis, vesicle transport and regulation of ciliary length and signaling. Direct binding of the membrane-associated guanylate kinase (MAGUK) DLG1 to the MAGUK-binding stalk domain of KIF13B relieves motor autoinhibition and promotes microtubule plus-end-directed cargo transport. Here, we characterize angiomotin (AMOT) isoform 2 (p80, referred to as Ap80) as a novel KIF13B interactor that promotes binding of another MAGUK, the polarity protein and Crumbs complex component PALS1, to KIF13B. Live-cell imaging analysis indicated that Ap80 is concentrated at and recruits PALS1 to the base of the primary cilium, but is not a cargo of KIF13B itself. Consistent with a ciliary function for Ap80, its depletion led to elongated primary cilia and reduced agonist-induced ciliary accumulation of SMO, a key component of the Hedgehog signaling pathway, whereas Ap80 overexpression caused ciliary shortening. Our results suggest that Ap80 activates KIF13B cargo binding at the base of the primary cilium to regulate ciliary length, composition and signaling., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
46. The cohesin acetylation cycle controls chromatin loop length through a PDS5A brake mechanism.
- Author
-
van Ruiten MS, van Gent D, Sedeño Cacciatore Á, Fauster A, Willems L, Hekkelman ML, Hoekman L, Altelaar M, Haarhuis JHI, Brummelkamp TR, de Wit E, and Rowland BD
- Subjects
- Acetylation, Chromatids metabolism, Chromatin, Histone Deacetylases genetics, Humans, Nuclear Proteins metabolism, Repressor Proteins genetics, Cohesins, Cell Cycle Proteins metabolism, Chromosomal Proteins, Non-Histone metabolism
- Abstract
Cohesin structures the genome through the formation of chromatin loops and by holding together the sister chromatids. The acetylation of cohesin's SMC3 subunit is a dynamic process that involves the acetyltransferase ESCO1 and deacetylase HDAC8. Here we show that this cohesin acetylation cycle controls the three-dimensional genome in human cells. ESCO1 restricts the length of chromatin loops, and of architectural stripes emanating from CTCF sites. HDAC8 conversely promotes the extension of such loops and stripes. This role in controlling loop length turns out to be distinct from the canonical role of cohesin acetylation that protects against WAPL-mediated DNA release. We reveal that acetylation controls the interaction of cohesin with PDS5A to restrict chromatin loop length. Our data support a model in which this PDS5A-bound state acts as a brake that enables the pausing and restart of loop enlargement. The cohesin acetylation cycle hereby provides punctuation in the process of genome folding., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
47. Ubiquitin ligase STUB1 destabilizes IFNγ-receptor complex to suppress tumor IFNγ signaling.
- Author
-
Apriamashvili G, Vredevoogd DW, Krijgsman O, Bleijerveld OB, Ligtenberg MA, de Bruijn B, Boshuizen J, Traets JJH, D'Empaire Altimari D, van Vliet A, Lin CP, Visser NL, Londino JD, Sanchez-Hodge R, Oswalt LE, Altinok S, Schisler JC, Altelaar M, and Peeper DS
- Subjects
- Humans, Immune Checkpoint Inhibitors, Signal Transduction, Ubiquitination, Interferon gamma Receptor, Interferon-gamma metabolism, Neoplasms immunology, Receptors, Interferon metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
The cytokine IFNγ differentially impacts on tumors upon immune checkpoint blockade (ICB). Despite our understanding of downstream signaling events, less is known about regulation of its receptor (IFNγ-R1). With an unbiased genome-wide CRISPR/Cas9 screen for critical regulators of IFNγ-R1 cell surface abundance, we identify STUB1 as an E3 ubiquitin ligase for IFNγ-R1 in complex with its signal-relaying kinase JAK1. STUB1 mediates ubiquitination-dependent proteasomal degradation of IFNγ-R1/JAK1 complex through IFNγ-R1
K285 and JAK1K249 . Conversely, STUB1 inactivation amplifies IFNγ signaling, sensitizing tumor cells to cytotoxic T cells in vitro. This is corroborated by an anticorrelation between STUB1 expression and IFNγ response in ICB-treated patients. Consistent with the context-dependent effects of IFNγ in vivo, anti-PD-1 response is increased in heterogenous tumors comprising both wildtype and STUB1-deficient cells, but not full STUB1 knockout tumors. These results uncover STUB1 as a critical regulator of IFNγ-R1, and highlight the context-dependency of STUB1-regulated IFNγ signaling for ICB outcome., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
48. Identification and characterization of Crumbs polarity complex proteins in Caenorhabditis elegans.
- Author
-
Castiglioni VG, Ramalho JJ, Kroll JR, Stucchi R, van Beuzekom H, Schmidt R, Altelaar M, and Boxem M
- Subjects
- Animals, Carrier Proteins genetics, Carrier Proteins metabolism, Cell Polarity genetics, Epithelial Cells cytology, Epithelial Cells metabolism, Epithelium metabolism, Caenorhabditis elegans genetics, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins genetics, Caenorhabditis elegans Proteins metabolism
- Abstract
Crumbs proteins are evolutionarily conserved transmembrane proteins with essential roles in promoting the formation of the apical domain in epithelial cells. The short intracellular tail of Crumbs proteins are known to interact with several proteins, including the scaffolding protein PALS1 (protein associated with LIN7, Stardust in Drosophila). PALS1 in turn binds to a second scaffolding protein PATJ (PALS1-associated tight junction protein) to form the core Crumbs/PALS1/PATJ complex. While essential roles in epithelial organization have been shown for Crumbs proteins in Drosophila and mammalian systems, the three Caenorhabditis elegans crumbs genes are dispensable for epithelial polarization and development. Here, we investigated the presence and function of PALS1 and PATJ orthologs in C. elegans. We identified MAGU-2 as the C. elegans ortholog of PALS1 and show that MAGU-2 interacts with all three Crumbs proteins and localizes to the apical membrane domain of intestinal epithelial cells in a Crumbs-dependent fashion. Similar to crumbs mutants, magu-2 deletion showed no epithelial polarity defects. We also identified MPZ-1 as a candidate ortholog of PATJ based on the physical interaction with MAGU-2 and sequence similarity with PATJ proteins. However, MPZ-1 is not broadly expressed in epithelial tissues and, therefore, not likely a core component of the C. elegans Crumbs complex. Finally, we show overexpression of the Crumbs proteins EAT-20 or CRB-3 can lead to apical membrane expansion in the intestine. Our results shed light on the composition of the C. elegans Crumbs complex and indicate that the role of Crumbs proteins in promoting apical domain formation is conserved., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
49. Tryptophan depletion results in tryptophan-to-phenylalanine substitutants.
- Author
-
Pataskar A, Champagne J, Nagel R, Kenski J, Laos M, Michaux J, Pak HS, Bleijerveld OB, Mordente K, Navarro JM, Blommaert N, Nielsen MM, Lovecchio D, Stone E, Georgiou G, de Gooijer MC, van Tellingen O, Altelaar M, Joosten RP, Perrakis A, Olweus J, Bassani-Sternberg M, Peeper DS, and Agami R
- Subjects
- Codon metabolism, Indoleamine-Pyrrole 2,3,-Dioxygenase genetics, Indoleamine-Pyrrole 2,3,-Dioxygenase metabolism, Interferon-gamma, Neoplasms immunology, Phenylalanine, T-Lymphocytes, Tryptophan Oxygenase genetics, Tryptophan Oxygenase metabolism, Tryptophan metabolism, Tryptophan-tRNA Ligase genetics, Tryptophan-tRNA Ligase metabolism
- Abstract
Activated T cells secrete interferon-γ, which triggers intracellular tryptophan shortage by upregulating the indoleamine 2,3-dioxygenase 1 (IDO1) enzyme
1-4 . Here we show that despite tryptophan depletion, in-frame protein synthesis continues across tryptophan codons. We identified tryptophan-to-phenylalanine codon reassignment (W>F) as the major event facilitating this process, and pinpointed tryptophanyl-tRNA synthetase (WARS1) as its source. We call these W>F peptides 'substitutants' to distinguish them from genetically encoded mutants. Using large-scale proteomics analyses, we demonstrate W>F substitutants to be highly abundant in multiple cancer types. W>F substitutants were enriched in tumours relative to matching adjacent normal tissues, and were associated with increased IDO1 expression, oncogenic signalling and the tumour-immune microenvironment. Functionally, W>F substitutants can impair protein activity, but also expand the landscape of antigens presented at the cell surface to activate T cell responses. Thus, substitutants are generated by an alternative decoding mechanism with potential effects on gene function and tumour immunoreactivity., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
50. Automated High-Throughput Method for the Fast, Robust, and Reproducible Enrichment of Newly Synthesized Proteins.
- Author
-
Vargas-Diaz D and Altelaar M
- Subjects
- Biotin chemistry, Click Chemistry, Peptides, Proteins analysis, Proteomics methods
- Abstract
A high-throughput method was developed for the automated enrichment of newly synthesized proteins (NSPs), which are labeled metabolically by substituting methionine with the "click-able" analogue azidohomoalanine (AHA). A suitable conjugate containing a dibenzocyclooctyne (DBCO) group allows the specific selection of NSPs by a fast 1 h click chemistry-based reaction with AHA. Through an automated pipetting platform, the samples are loaded into streptavidin cartridges for the selective binding of the NSPs by means of a biotin bait contained in the conjugate. The enriched proteins are eluted by a reproducible chemical cleavage of the 4,4-dimethyl-2,6-dioxocyclohexylidene (Dde) group in the conjugate, which increases selectivity. The NSPs can be collected and digested in the same well plate, and the resulting peptides can be subsequently loaded for automated cleanup, followed by mass spectrometry analysis. The proposed automated method allows for the robust and effective enrichment of samples in 96-well plates in a period of 3 h. Our developed enrichment method was comprehensively evaluated and then applied to the proteomics analysis of the melanoma A375 cell secretome, after treatment with the cytokines interferon α (IFN-α) and γ (IFN-γ), resulting in the quantification of 283 and 263 proteins, respectively, revealing intricate tumor growth-supportive and -suppressive effects.
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.