28 results on '"Krotenberg Garcia A."'
Search Results
2. Cell competition in primary and metastatic colorectal cancer
- Author
-
Merel Elise van Luyk, Ana Krotenberg Garcia, Maria Lamprou, and Saskia Jacoba Elisabeth Suijkerbuijk
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Adult tissues set the scene for a continuous battle between cells, where a comparison of cellular fitness results in the elimination of weaker “loser” cells. This phenomenon, named cell competition, is beneficial for tissue integrity and homeostasis. In fact, cell competition plays a crucial role in tumor suppression, through elimination of early malignant cells, as part of Epithelial Defense Against Cancer. However, it is increasingly apparent that cell competition doubles as a tumor-promoting mechanism. The comparative nature of cell competition means that mutational background, proliferation rate and polarity all factor in to determine the outcome of these processes. In this review, we explore the intricate and context-dependent involvement of cell competition in homeostasis and regeneration, as well as during initiation and progression of primary and metastasized colorectal cancer. We provide a comprehensive overview of molecular and cellular mechanisms governing cell competition and its parallels with regeneration.
- Published
- 2024
- Full Text
- View/download PDF
3. Subtype-specific kinase dependency regulates growth and metastasis of poor-prognosis mesenchymal colorectal cancer
- Author
-
Buikhuisen, Joyce Y., Gomez Barila, Patricia M., Cameron, Kate, Suijkerbuijk, Saskia J. E., Lieftink, Cor, di Franco, Simone, Krotenberg Garcia, Ana, Uceda Castro, Rebeca, Lenos, Kristiaan J., Nijman, Lisanne E., Torang, Arezo, Longobardi, Ciro, de Jong, Joan H., Dekker, Daniëlle, Stassi, Giorgio, Vermeulen, Louis, Beijersbergen, Roderick L., van Rheenen, Jacco, Huveneers, Stephan, and Medema, Jan Paul
- Published
- 2023
- Full Text
- View/download PDF
4. Cell competition promotes metastatic intestinal cancer through a multistage process
- Author
-
Ana Krotenberg Garcia, Mario Ledesma-Terrón, Maria Lamprou, Joyce Vriend, Merel Elise van Luyk, and Saskia Jacoba Elisabeth Suijkerbuijk
- Subjects
Microenvironment ,Cell biology ,Cancer ,Science - Abstract
Summary: Cell competition plays an instrumental role in quality control during tissue development and homeostasis. Nevertheless, cancer cells can exploit this process for their own proliferative advantage. In our study, we generated mixed murine organoids and microtissues to explore the impact of cell competition on liver metastasis. Unlike competition at the primary site, the initial effect on liver progenitor cells does not involve the induction of apoptosis. Instead, metastatic competition manifests as a multistage process. Initially, liver progenitors undergo compaction, which is followed by cell-cycle arrest, ultimately forcing differentiation. Subsequently, the newly differentiated liver cells exhibit reduced cellular fitness, rendering them more susceptible to outcompetition by intestinal cancer cells. Notably, cancer cells leverage different interactions with different epithelial populations in the liver, using them as scaffolds to facilitate their growth. Consequently, tissue-specific mechanisms of cell competition are fundamental in driving metastatic intestinal cancer.
- Published
- 2024
- Full Text
- View/download PDF
5. Cell competition promotes metastatic intestinal cancer through a multistage process
- Author
-
Krotenberg Garcia, Ana, Ledesma-Terrón, Mario, Lamprou, Maria, Vriend, Joyce, van Luyk, Merel Elise, and Suijkerbuijk, Saskia Jacoba Elisabeth
- Published
- 2024
- Full Text
- View/download PDF
6. Subtype-specific kinase dependency regulates growth and metastasis of poor-prognosis mesenchymal colorectal cancer
- Author
-
Joyce Y. Buikhuisen, Patricia M. Gomez Barila, Kate Cameron, Saskia J. E. Suijkerbuijk, Cor Lieftink, Simone di Franco, Ana Krotenberg Garcia, Rebeca Uceda Castro, Kristiaan J. Lenos, Lisanne E. Nijman, Arezo Torang, Ciro Longobardi, Joan H. de Jong, Daniëlle Dekker, Giorgio Stassi, Louis Vermeulen, Roderick L. Beijersbergen, Jacco van Rheenen, Stephan Huveneers, and Jan Paul Medema
- Subjects
Colorectal cancer ,Epithelial-mesenchymal transition ,Metastasis ,Cellular attachment ,PAK family ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Colorectal cancer (CRC) can be divided into four consensus molecular subtypes (CMS), each with distinct biological features. CMS4 is associated with epithelial-mesenchymal transition and stromal infiltration (Guinney et al., Nat Med 21:1350–6, 2015; Linnekamp et al., Cell Death Differ 25:616–33, 2018), whereas clinically it is characterized by lower responses to adjuvant therapy, higher incidence of metastatic spreading and hence dismal prognosis (Buikhuisen et al., Oncogenesis 9:66, 2020). Methods To understand the biology of the mesenchymal subtype and unveil specific vulnerabilities, a large CRISPR-Cas9 drop-out screen was performed on 14 subtyped CRC cell lines to uncover essential kinases in all CMSs. Dependency of CMS4 cells on p21-activated kinase 2 (PAK2) was validated in independent 2D and 3D in vitro cultures and in vivo models assessing primary and metastatic outgrowth in liver and peritoneum. TIRF microscopy was used to uncover actin cytoskeleton dynamics and focal adhesion localization upon PAK2 loss. Subsequent functional assays were performed to determine altered growth and invasion patterns. Results PAK2 was identified as a key kinase uniquely required for growth of the mesenchymal subtype CMS4, both in vitro and in vivo. PAK2 plays an important role in cellular attachment and cytoskeletal rearrangements (Coniglio et al., Mol Cell Biol 28:4162–72, 2008; Grebenova et al., Sci Rep 9:17171, 2019). In agreement, deletion or inhibition of PAK2 impaired actin cytoskeleton dynamics in CMS4 cells and, as a consequence, significantly reduced invasive capacity, while it was dispensable for CMS2 cells. Clinical relevance of these findings was supported by the observation that deletion of PAK2 from CMS4 cells prevented metastatic spreading in vivo. Moreover, growth in a model for peritoneal metastasis was hampered when CMS4 tumor cells were deficient for PAK2. Conclusion Our data reveal a unique dependency of mesenchymal CRC and provide a rationale for PAK2 inhibition to target this aggressive subgroup of colorectal cancer.
- Published
- 2023
- Full Text
- View/download PDF
7. Ribosome impairment regulates intestinal stem cell identity via ZAKɑ activation
- Author
-
Joana Silva, Ferhat Alkan, Sofia Ramalho, Goda Snieckute, Stefan Prekovic, Ana Krotenberg Garcia, Santiago Hernández-Pérez, Rob van der Kammen, Danielle Barnum, Liesbeth Hoekman, Maarten Altelaar, Wilbert Zwart, Saskia Jacoba Elisabeth Suijkerbuijk, Simon Bekker-Jensen, and William James Faller
- Subjects
Science - Abstract
Intestinal stem cells are responsible for replenishing cells within the high-turnover intestinal epithelium. Here they show that ribosome dynamics affect intestinal stem cell identity through a mechanism that is triggered by changes in nutrient availability.
- Published
- 2022
- Full Text
- View/download PDF
8. Integrin α3β1 Is a Key Regulator of Several Protumorigenic Pathways during Skin Carcinogenesis
- Author
-
Ramovs, Veronika, Krotenberg Garcia, Ana, Kreft, Maaike, and Sonnenberg, Arnoud
- Published
- 2021
- Full Text
- View/download PDF
9. Cell competition promotes metastatic intestinal cancer through a multistage process
- Author
-
Krotenberg Garcia, A, Ledesma-Terrón, M, Lamprou, M, Vriend, J, van Luyk, ME, Suijkerbuijk, SJE, Krotenberg Garcia, A, Ledesma-Terrón, M, Lamprou, M, Vriend, J, van Luyk, ME, and Suijkerbuijk, SJE
- Abstract
Cell competition plays an instrumental role in quality control during tissue development and homeostasis. Nevertheless, cancer cells can exploit this process for their own proliferative advantage. In our study, we generated mixed murine organoids and microtissues to explore the impact of cell competition on liver metastasis. Unlike competition at the primary site, the initial effect on liver progenitor cells does not involve the induction of apoptosis. Instead, metastatic competition manifests as a multistage process. Initially, liver progenitors undergo compaction, which is followed by cell-cycle arrest, ultimately forcing differentiation. Subsequently, the newly differentiated liver cells exhibit reduced cellular fitness, rendering them more susceptible to outcompetition by intestinal cancer cells. Notably, cancer cells leverage different interactions with different epithelial populations in the liver, using them as scaffolds to facilitate their growth. Consequently, tissue-specific mechanisms of cell competition are fundamental in driving metastatic intestinal cancer.
- Published
- 2024
10. The role of cell competition in intestinal cancer: from primary tumor to liver metastasis
- Author
-
Krotenberg Garcia, Ana and Krotenberg Garcia, Ana
- Abstract
The significance of cell competition in disease, particularly in cancer progression, has become increasingly apparent. Throughout tumorigenesis, cell-to-cell interactions play a key role in enabling cells to assess the fitness of their neighboring cells. This leads to the establishment of whether mutated cells will be outcompeted by healthy tissue, employing competition as a tumor suppression mechanism. Conversely, aberrant cells may become “supercompetitors,” and win the battle with healthy cells. In this thesis, we delve into the interactions between intestinal wild-type and cancer cells during primary tumor and metastasis formation. Chapter 2 outlines the development of a co-culture system that enables the investigation of direct cellular interactions between two epithelial cell types within the same organoid. This model allows the dissection of interactions between cells with a differential fitness based on both direct contact and short-range paracrine signaling. Our focus lies specifically on the interaction between intestinal wild-type and cancer cells, and it excludes the influence of non-epithelial cells such as those of the immune system. In Chapter 3, we demonstrate that the presence of intestinal cancer cells induces wild-type intestinal cells to revert to a fetal-like state. This reversion is accompanied by increased JNK activation and results in the active elimination of the wild-type population. Cancer cells exploit this competition to enhance their own growth. Chapter 4 explores cell competition in a metastatic environment. We observe that intestinal cancer cells cause compaction of liver progenitor cells, leading to forced differentiation that is characterized by a cell-cycle arrest. While active elimination of liver progenitors is not observed, cancer cells can exploit these competitive interactions to increase their proliferation. In microtissues, competitive behavior of cancer cells manifests differently, here wild-type hepatocyte-like cells are u
- Published
- 2024
11. The role of cell competition in intestinal cancer: from primary tumor to liver metastasis
- Author
-
Sub Developmental Biology, Developmental Biology, van den Heuvel, Sander, van Rheenen, Jacobus, Suijkerbuijk, Saskia, Krotenberg Garcia, Ana, Sub Developmental Biology, Developmental Biology, van den Heuvel, Sander, van Rheenen, Jacobus, Suijkerbuijk, Saskia, and Krotenberg Garcia, Ana
- Published
- 2024
12. Generation of mixed murine organoids to model cellular interactions
- Author
-
Ana Krotenberg Garcia, Jacco van Rheenen, and Saskia Jacoba Elisabeth Suijkerbuijk
- Subjects
Cancer ,Cell Biology ,Cell culture ,Microscopy ,Organoids ,Science (General) ,Q1-390 - Abstract
Summary: Cell competition is a mechanism of interaction that dictates cell selection based on differences in cellular fitness. We designed a protocol to generate mixed murine organoids and enteroid monolayers used to study such complex cellular interactions in a mammalian system. This protocol is dedicated to follow the behavior of different cell populations over time, using (time-lapse) microscopy or transcriptome/proteome analysis.For complete details on the use and execution of this protocol, please refer to Krotenberg Garcia et al. (2021).
- Published
- 2021
- Full Text
- View/download PDF
13. Mechanical regulation of cell fate transitions underlying colorectal cancer metastasis formation
- Author
-
van der Net, Mirjam C, primary, Vliem, Marjolein J, additional, Kemp, Lars J S, additional, Perez-Gonzalez, Carlos, additional, Strating, Esther A, additional, Krotenberg-Garcia, Ana, additional, Houtekamer, Ronja M, additional, van den Anker, Karen B, additional, Monster, Jooske L, additional, Snippert, Hugo J G, additional, Khalil, Antoine A, additional, van Rheenen, Jacco, additional, Suijkerbuijk, Saskia JE, additional, Kranenburg, Onno, additional, Matic Vignjevic, Danijela, additional, and Gloerich, Martijn, additional
- Published
- 2023
- Full Text
- View/download PDF
14. Cell competition promotes metastatic intestinal cancer through a multistage process
- Author
-
Krotenberg Garcia, Ana, primary, Ledesma-Terron, Mario, additional, Vriend, Joyce, additional, van Luyk, Merel E, additional, and Suijkerbuijk, Saskia JE, additional
- Published
- 2023
- Full Text
- View/download PDF
15. Mechanical regulation of cell fate transitions underlying colorectal cancer metastasis formation
- Author
-
Sub Developmental Biology, Developmental Biology, Net, Mirjam van der, Vliem, Marjolein J, Kemp, Lars, Perez-Gonzalez, Carlos, Strating, Esther A, Krotenberg-Garcia, Ana, Houtekamer, Ronja, Anker, Karen B van den, Monster, Jooske, Snippert, Hugo, Khalil, Antoine A., Rheenen, Jacco van, Suijkerbuijk, Saskia J E, Kranenburg, Onno, vignjevic, Danijela Matic, Gloerich, Martijn, Sub Developmental Biology, Developmental Biology, Net, Mirjam van der, Vliem, Marjolein J, Kemp, Lars, Perez-Gonzalez, Carlos, Strating, Esther A, Krotenberg-Garcia, Ana, Houtekamer, Ronja, Anker, Karen B van den, Monster, Jooske, Snippert, Hugo, Khalil, Antoine A., Rheenen, Jacco van, Suijkerbuijk, Saskia J E, Kranenburg, Onno, vignjevic, Danijela Matic, and Gloerich, Martijn
- Published
- 2023
16. Additional file 1 of Subtype-specific kinase dependency regulates growth and metastasis of poor-prognosis mesenchymal colorectal cancer
- Author
-
Buikhuisen, Joyce Y., Gomez Barila, Patricia M., Cameron, Kate, Suijkerbuijk, Saskia J. E., Lieftink, Cor, di Franco, Simone, Krotenberg Garcia, Ana, Uceda Castro, Rebeca, Lenos, Kristiaan J., Nijman, Lisanne E., Torang, Arezo, Longobardi, Ciro, de Jong, Joan H., Dekker, Daniëlle, Stassi, Giorgio, Vermeulen, Louis, Beijersbergen, Roderick L., van Rheenen, Jacco, Huveneers, Stephan, and Medema, Jan Paul
- Abstract
Additional file 1: Supplementary Fig. S1. Validation of PAK2 as an essential kinase for CMS4 cell lines. A, PAK1–3 mRNA expression levels in a panel of 28 CRC cell lines, also including those used for the drop-out screen, as determined by quantitative PCR. Of note: diamond for PAK3 located on x-axis indicates no mRNA could be detected in this sample. B, C, 2Log mRNA expression levels of PAK4–6 in CRC cell lines (B) and tumors (C), determined by microarray or RNA sequencing. D, Western blot for PAK1 protein expression in HT55 & SW948 (CMS2) and HuTu-80 & MDST8 (CMS4). 2,2,2-Trichloroethanol (2,2,2TCE) signal (excerpt taken around 60 kDa region) indicates amount of protein loaded per cell line. Numbers on the left represent molecular weight in kDa according to a stained protein ladder loaded in the gel. Representative blot of N = 3 is displayed. E, F, PAK2 (E) and PAK1 (F) gene expression levels in cells expressing non-silencing (NS) or PAK2 targeting shRNA (PAK2-KD), as determined by qPCR. Expression was normalized to NS condition, N = 3, mean + S.D. is plotted. G, PAK2 gene expression levels in cells expressing non-silencing (NS) or PAK2 targeting shRNA (PAK2-KD), as determined by qPCR in CMS4 cell lines not included in the initial screen (CaR-1, LS123) and a CMS4 line in which PAK2 was not a significant drop-out in the initial screen (Colo320-HSR). H, Relative contribution of PAK2 KD cells to co-culture in competition assay set-up, over time, in additional CMS4 cell lines. Contribution of shRNA-turboRFP expressing cells was first normalized to contribution on day 0 post sort, and subsequently calculated relative to contribution of NS shRNA-turboRFP expressing cells at each individual time-point. N ≥ 2, mean + s.e.m. is plotted. Supplementary Fig. S2. PAK2 loss alters cell morphology, not induction of apoptosis, of CMS4 cell lines. A, Validation of successful PAK2 knock-out (KO) in OUMS-23 CRISPR-Cas9-edited single cell knock-out clones. WT = wildtype. Numbers on the left represent molecular weight in kDa according to a stained protein ladder loaded in the gel. B, Phase contrast images of OUMS-23 WT and KO cells 7 days post-plating. Region of interest (ROI) selected for lower panels is indicated in white dashed box. C, D, Outgrowth of WT and PAK2-KO cells plated on plastic (C) or Matrigel (D) relative to day 0, as measured 4 days post-plating by CellTiter Glo. Representative experiment of N = 3 is shown. Data plotted is mean + S.D. N.S. = not significant, ** = P < 0.005, *** = P < 0.001, **** = P < 0.0001 in two-tailed, Welch-corrected Student’s T-test. E, F, G, Relative outgrowth of cells after 3 days of treatment with different concentrations of Oxaliplatin (E), 5-FU (F) and Paclitaxel (G) measured using CellTiter Glo and normalized to DMSO treated control relative to a day 0 baseline measurement. Data plotted is mean ± S.D. Representative experiment of N = 3 is shown. H, Percentage of apoptotic cells present 48 hours post plating as assessed by Nicoletti assay. Bar graphs depict mean + s.e.m., N = 3, n.s. = not significant as calculated by two-tailed, Welch-corrected Student’s T-test. I, Phase contrast images of CMS2 (HT55) and CMS4 (HuTu-80, MDST8) WT and KO cells 5 days post-plating. Scale bar represents 50 μm. Supplementary Fig. S3. PAK2 deficiency affects actin cytoskeleton turnover and focal adhesion localization. A, ARHGEF7 (βPIX) gene expression levels in cells expressing non-silencing (NS) or ARHGEF7 (βPIX) targeting shRNAs (Sh-1, Sh-2), as determined by qPCR. Expression was normalized to NS condition, N = 3, mean + s.d. is plotted. B, C, Region of interest stills (Fig. 4D) from time-lapse TIRF microscopy at indicated time points (min = minutes) of cell lines transduced with Lifeact-GFP (green) and paxillin-mCherry (magenta). Scale bars indicate 5 μm. See corresponding Supplementary Movies 5and 6, available online, for the full time-lapse recording. D, E, Heatmaps show actin cytoskeleton dynamics over 60 minutes for HuTu-80 (D) and MDST8 (E) in a unique color per time frame. Note the rapid emergence and disappearance of filopodia in the WT condition and relative stability of filopodia over time in PAK2-KO cells. Supplementary Fig. S4. Metastatic colonization and secondary spread is strongly reduced by PAK2 loss. A, PAK2 gene expression levels in primary cultures expressing non-silencing (NS) or PAK2 targeting shRNA (PAK2-KD), as determined by qPCR. B, Relative outgrowth of NS or PAK2-KD primary cell lines plated on low-adherent culture plates relative to day 0, as measured 4 days post-plating by CellTiter Glo.. Data plotted is mean ± S.D. Representative experiment of N = 3 is shown. C, D, Quantification (C) and representative images (D) of Migration Transwell experiments. Bar plots display relative area percentage covered by cells (purple) in Transwell image. Data plotted is mean ± S.D. Values representing N = 2 are plotted. E, Representative images of liver and lung sections stained for human Ku80 (DAB, indicated in brown), derived from two mice per condition. Along with the other Ku80-stained sections, these images were the source for quantification as included in Fig. 5G, I. F, Macroscopic images of tumor burden in full livers and lungs of mice injected with a 50–50% mixture of HuTu-80 WT-Dendra (green) and PAK2-KO-tdTomato cells (magenta). Pictures were taken shortly after mice were sacrificed.
- Published
- 2023
- Full Text
- View/download PDF
17. Human-iPSC-Derived Cardiac Stromal Cells Enhance Maturation in 3D Cardiac Microtissues and Reveal Non-cardiomyocyte Contributions to Heart Disease
- Author
-
Giacomelli, E, Meraviglia, V, Campostrini, G, Cochrane, A, Cao, X, van Helden, R, Krotenberg Garcia, A, Mircea, M, Kostidis, S, Davis, R, van Meer, B, Jost, C, Koster, A, Mei, H, Miguez, D, Mulder, A, Ledesma-Terron, M, Pompilio, G, Sala, L, Salvatori, D, Slieker, R, Sommariva, E, de Vries, A, Giera, M, Semrau, S, Tertoolen, L, Orlova, V, Bellin, M, Mummery, C, Giacomelli E., Meraviglia V., Campostrini G., Cochrane A., Cao X., van Helden R. W. J., Krotenberg Garcia A., Mircea M., Kostidis S., Davis R. P., van Meer B. J., Jost C. R., Koster A. J., Mei H., Miguez D. G., Mulder A. A., Ledesma-Terron M., Pompilio G., Sala L., Salvatori D. C. F., Slieker R. C., Sommariva E., de Vries A. A. F., Giera M., Semrau S., Tertoolen L. G. J., Orlova V. V., Bellin M., Mummery C. L., Giacomelli, E, Meraviglia, V, Campostrini, G, Cochrane, A, Cao, X, van Helden, R, Krotenberg Garcia, A, Mircea, M, Kostidis, S, Davis, R, van Meer, B, Jost, C, Koster, A, Mei, H, Miguez, D, Mulder, A, Ledesma-Terron, M, Pompilio, G, Sala, L, Salvatori, D, Slieker, R, Sommariva, E, de Vries, A, Giera, M, Semrau, S, Tertoolen, L, Orlova, V, Bellin, M, Mummery, C, Giacomelli E., Meraviglia V., Campostrini G., Cochrane A., Cao X., van Helden R. W. J., Krotenberg Garcia A., Mircea M., Kostidis S., Davis R. P., van Meer B. J., Jost C. R., Koster A. J., Mei H., Miguez D. G., Mulder A. A., Ledesma-Terron M., Pompilio G., Sala L., Salvatori D. C. F., Slieker R. C., Sommariva E., de Vries A. A. F., Giera M., Semrau S., Tertoolen L. G. J., Orlova V. V., Bellin M., and Mummery C. L.
- Abstract
Cardiomyocytes (CMs) from human induced pluripotent stem cells (hiPSCs) are functionally immature, but this is improved by incorporation into engineered tissues or forced contraction. Here, we showed that tri-cellular combinations of hiPSC-derived CMs, cardiac fibroblasts (CFs), and cardiac endothelial cells also enhance maturation in easily constructed, scaffold-free, three-dimensional microtissues (MTs). hiPSC-CMs in MTs with CFs showed improved sarcomeric structures with T-tubules, enhanced contractility, and mitochondrial respiration and were electrophysiologically more mature than MTs without CFs. Interactions mediating maturation included coupling between hiPSC-CMs and CFs through connexin 43 (CX43) gap junctions and increased intracellular cyclic AMP (cAMP). Scaled production of thousands of hiPSC-MTs was highly reproducible across lines and differentiated cell batches. MTs containing healthy-control hiPSC-CMs but hiPSC-CFs from patients with arrhythmogenic cardiomyopathy strikingly recapitulated features of the disease. Our MT model is thus a simple and versatile platform for modeling multicellular cardiac diseases that will facilitate industry and academic engagement in high-throughput molecular screening.
- Published
- 2020
18. Blinded, Multicenter Evaluation of Drug-induced Changes in Contractility Using Human-induced Pluripotent Stem Cell-derived Cardiomyocytes
- Author
-
Nurul A N Mohd Yusof, Christine L. Mummery, Leon G.J. Tertoolen, Jessica Nebel, Peter Clements, Eric I. Rossman, Umber Saleem, Ana Krotenberg Garcia, Kate Harris, Maria L. H. Vlaming, Karen McGlynn, Godfrey L. Smith, Xiaoping Xu, Puspita A Katili, Tessa de Korte, Francis L. Burton, Berend J. van Meer, Ingra Mannhardt, Arne Hansen, Chris Denning, Anthony Bahinski, Thomas Eschenhagen, and Applied Stem Cell Technologies
- Subjects
0301 basic medicine ,Drug ,Inotrope ,CRACK-IT project ,medicine.medical_specialty ,Contraction (grammar) ,media_common.quotation_subject ,Induced Pluripotent Stem Cells ,Alternatives to animal testing ,cardiomyocytes ,predictive toxicology ,030204 cardiovascular system & hematology ,Toxicology ,contractility ,Contractility ,03 medical and health sciences ,alternatives to animal testing ,0302 clinical medicine ,Internal medicine ,Emerging Technologies, Methods, and Models ,safety pharmacology ,medicine ,human-induced pluripotent stem cells ,Animals ,Humans ,Myocytes, Cardiac ,Induced pluripotent stem cell ,media_common ,Dose-Response Relationship, Drug ,business.industry ,Safety pharmacology ,inotropy ,electrophysiology ,Electrophysiology ,030104 developmental biology ,Pharmaceutical Preparations ,Cardiology ,business - Abstract
Animal models are 78% accurate in determining whether drugs will alter contractility of the human heart. To evaluate the suitability of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) for predictive safety pharmacology, we quantified changes in contractility, voltage, and/or Ca2+ handling in 2D monolayers or 3D engineered heart tissues (EHTs). Protocols were unified via a drug training set, allowing subsequent blinded multicenter evaluation of drugs with known positive, negative, or neutral inotropic effects. Accuracy ranged from 44% to 85% across the platform-cell configurations, indicating the need to refine test conditions. This was achieved by adopting approaches to reduce signal-to-noise ratio, reduce spontaneous beat rate to ≤ 1 Hz or enable chronic testing, improving accuracy to 85% for monolayers and 93% for EHTs. Contraction amplitude was a good predictor of negative inotropes across all the platform-cell configurations and of positive inotropes in the 3D EHTs. Although contraction- and relaxation-time provided confirmatory readouts forpositive inotropes in 3D EHTs, these parameters typically served as the primary source of predictivity in 2D. The reliance of these “secondary” parameters to inotropy in the 2D systems was not automatically intuitive and may be a quirk of hiPSC-CMs, hence require adaptations in interpreting the data from this model system. Of the platform-cell configurations, responses in EHTs aligned most closely to the free therapeutic plasma concentration. This study adds to the notion that hiPSC-CMs could add value to drug safety evaluation.
- Published
- 2020
19. Generation of mixed murine organoids to model cellular interactions
- Author
-
Krotenberg Garcia, Ana, primary, van Rheenen, Jacco, additional, and Suijkerbuijk, Saskia Jacoba Elisabeth, additional
- Published
- 2021
- Full Text
- View/download PDF
20. Active elimination of intestinal cells drives oncogenic growth in organoids
- Author
-
Krotenberg Garcia, Ana, primary, Fumagalli, Arianna, additional, Le, Huy Quang, additional, Jackstadt, Rene, additional, Lannagan, Tamsin Rosemary Margaret, additional, Sansom, Owen James, additional, van Rheenen, Jacco, additional, and Suijkerbuijk, Saskia Jacoba Elisabeth, additional
- Published
- 2021
- Full Text
- View/download PDF
21. Active elimination of intestinal cells drives oncogenic growth in organoids
- Author
-
Suijkerbuijk, Krotenberg Garcia, Sansom, Fumagalli, Quang Le, and van Rheenen
- Subjects
education.field_of_study ,Programmed cell death ,Chemistry ,Cell ,Population ,Cell fate determination ,Cell biology ,medicine.anatomical_structure ,Signalling ,Cancer cell ,medicine ,Organoid ,education ,Homeostasis - Abstract
Competitive cell-interactions play a crucial role in quality control during development and homeostasis. Here we show that cancer cells use such interactions to actively eliminate wild-type intestine cells in enteroid monolayers and organoids. This apoptosis-dependent process boosts proliferation of intestinal cancer cells. The remaining wild-type population activates markers of primitive epithelia and transits to a fetal-like state. Prevention of this cell fate transition avoids elimination of wild-type cells and, importantly, limits the proliferation of cancer cells. JNK signalling is activated in competing cells and is required for cell fate change and elimination of wild-type cells. Thus, cell competition drives growth of cancer cells by active out-competition of wild-type cells through forced cell death and cell fate change in a JNK dependent manner.
- Published
- 2020
22. Active elimination of intestinal cells drives oncogenic growth in organoids
- Author
-
Owen J. Sansom, Huy Quang Le, Jacco van Rheenen, Arianna Fumagalli, Rene Jackstadt, Tamsin Rosemary Margaret Lannagan, Saskia J.E. Suijkerbuijk, and Ana Krotenberg Garcia
- Subjects
Male ,Programmed cell death ,Carcinogenesis ,MAP Kinase Signaling System ,Population ,Cell ,Apoptosis ,Mice, Transgenic ,General Biochemistry, Genetics and Molecular Biology ,Article ,Receptors, G-Protein-Coupled ,Fetus ,Cell Line, Tumor ,Organoid ,medicine ,Animals ,cancer ,education ,education.field_of_study ,Kinase ,Chemistry ,Stem Cells ,Cancer ,medicine.disease ,Cell biology ,Intestines ,Mice, Inbred C57BL ,Organoids ,medicine.anatomical_structure ,Cell Competition ,Cancer cell ,fetal-like ,JNK ,small intestine ,Homeostasis - Abstract
Summary Competitive cell interactions play a crucial role in quality control during development and homeostasis. Here, we show that cancer cells use such interactions to actively eliminate wild-type intestine cells in enteroid monolayers and organoids. This apoptosis-dependent process boosts proliferation of intestinal cancer cells. The remaining wild-type population activates markers of primitive epithelia and transits to a fetal-like state. Prevention of this cell-state transition avoids elimination of wild-type cells and, importantly, limits the proliferation of cancer cells. Jun N-terminal kinase (JNK) signaling is activated in competing cells and is required for cell-state change and elimination of wild-type cells. Thus, cell competition drives growth of cancer cells by active out-competition of wild-type cells through forced cell death and cell-state change in a JNK-dependent manner., Graphical abstract, Highlights • In organoids, wild-type intestine cells are actively eliminated by cancer cells • Cell competition boosts proliferation of intestinal cancer cells • Remaining wild-type cells adopt a fetal-like state • JNK signaling in wild-type cells drives cell competition, Interactions between cells helps coordinated decision making in tissues. Krotenberg Garcia et al. show that intestinal cancer cells use competitive cell interactions to actively eliminate surrounding wild-type cells. These interactions drive oncogenic growth in organoids.
- Published
- 2020
23. Integrin α3β1 Is a Key Regulator of Several Protumorigenic Pathways during Skin Carcinogenesis
- Author
-
Veronika Ramovs, Ana Krotenberg Garcia, Arnoud Sonnenberg, and Maaike Kreft
- Subjects
0301 basic medicine ,Keratinocytes ,Skin Neoplasms ,Cell Survival ,9,10-Dimethyl-1,2-benzanthracene ,Integrin ,DMBA ,Dermatology ,Tetraspanin 24 ,Biochemistry ,Cell Line ,Focal adhesion ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Spheroids, Cellular ,Cell Adhesion ,Animals ,Humans ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,biology ,Chemistry ,Integrin alpha3beta1 ,Cell Biology ,Neoplasms, Experimental ,Cell biology ,030104 developmental biology ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,biology.protein ,STAT protein ,Carcinogens ,Tetradecanoylphorbol Acetate ,Signal transduction ,Epidermis ,Cell Adhesion Molecules ,Proto-oncogene tyrosine-protein kinase Src ,Signal Transduction - Abstract
Integrin α3β1 plays a crucial role in tumor formation in the two-stage chemical carcinogenesis model (DMBA and TPA treatment). However, the mechanisms whereby the expression of α3β1 influences key oncogenic drivers of this established model are not known yet. Using an in vivo mouse model with epidermal deletion of α3β1 and in vitro Matrigel cultures of transformed keratinocytes, we demonstrate the central role of α3β1 in promoting the activation of several protumorigenic signaling pathways during the initiation of DMBA/TPA‒driven tumorigenesis. In transformed keratinocytes, α3β1-mediated focal adhesion kinase/Src activation leads to in vitro growth of spheroids and to strong Akt and STAT 3 activation when the α3β1-binding partner tetraspanin CD151 is present to stabilize cell‒cell adhesion and promote Smad2 phosphorylation. Remarkably, α3β1 and CD151 can support Akt and STAT 3 activity independently of α3β1 ligation by laminin-332 and as such control the essential survival signals required for suprabasal keratin-10 expression during keratinocyte differentiation. These data demonstrate that α3β1 together with CD151 regulate the signaling pathways that control the survival of differentiating keratinocytes and provide a mechanistic understanding of the essential role of α3β1 in early stages of skin cancer development.
- Published
- 2020
24. Human iPSC-derived cardiac stromal cells enhance maturation in 3D cardiac microtissues and reveal non-cardiomyocyte contributions to heart disease
- Author
-
Elisa, Giacomelli, Viviana, Meraviglia, Giulia, Campostrini, Amy, Cochrane, Cao, Xu, van Helden, Ruben W. J., Ana Krotenberg Garcia, Maria, Mircea, Sarantos, Kostidis, Davis, Richard P., van Meer, Berend J., Jost, Carolina R., Koster, Abraham J., Hailiang, Mei, Míguez, David G., Mulder, Aat A., Mario, Ledesma-Terrón, Giulio, Pompilio, Luca, Sala, Salvatori, Daniela C. F., Slieker, Roderick C., Elena, Sommariva, de Vries, Antoine A. F., Martin, Giera, Stefan, Semrau, Tertoolen, Leon G. J., Orlova, Valeria V., Bellin, Milena, and Mummery, Christine L.
- Published
- 2020
25. Human-iPSC-Derived Cardiac Stromal Cells Enhance Maturation in 3D Cardiac Microtissues and Reveal Non-cardiomyocyte Contributions to Heart Disease
- Author
-
Giacomelli, Elisa, primary, Meraviglia, Viviana, additional, Campostrini, Giulia, additional, Cochrane, Amy, additional, Cao, Xu, additional, van Helden, Ruben W.J., additional, Krotenberg Garcia, Ana, additional, Mircea, Maria, additional, Kostidis, Sarantos, additional, Davis, Richard P., additional, van Meer, Berend J., additional, Jost, Carolina R., additional, Koster, Abraham J., additional, Mei, Hailiang, additional, Míguez, David G., additional, Mulder, Aat A., additional, Ledesma-Terrón, Mario, additional, Pompilio, Giulio, additional, Sala, Luca, additional, Salvatori, Daniela C.F., additional, Slieker, Roderick C., additional, Sommariva, Elena, additional, de Vries, Antoine A.F., additional, Giera, Martin, additional, Semrau, Stefan, additional, Tertoolen, Leon G.J., additional, Orlova, Valeria V., additional, Bellin, Milena, additional, and Mummery, Christine L., additional
- Published
- 2020
- Full Text
- View/download PDF
26. Integrin α3β1 in hair bulge stem cells modulates CCN2 expression and promotes skin tumorigenesis
- Author
-
Ramovs, Veronika, primary, Krotenberg Garcia, Ana, additional, Song, Ji-Ying, additional, de Rink, Iris, additional, Kreft, Maaike, additional, Goldschmeding, Roel, additional, and Sonnenberg, Arnoud, additional
- Published
- 2020
- Full Text
- View/download PDF
27. Integrin α3β1 in hair bulge stem cells modulates CCN2 expression and promotes skin tumorigenesis
- Author
-
Veronika Ramovs, Roel Goldschmeding, Ana Krotenberg Garcia, Iris de Rink, Maaike Kreft, Ji-Ying Song, and Arnoud Sonnenberg
- Subjects
Keratinocytes ,0301 basic medicine ,Skin Neoplasms ,Health, Toxicology and Mutagenesis ,medicine.medical_treatment ,Integrin ,Fluorescent Antibody Technique ,Gene Expression ,DMBA ,Connective tissue ,Plant Science ,medicine.disease_cause ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Immunophenotyping ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Research Articles ,Neoplasm Staging ,Mice, Knockout ,integumentary system ,Ecology ,biology ,Chemistry ,Stem Cells ,Growth factor ,Matricellular protein ,Connective Tissue Growth Factor ,Integrin alpha3beta1 ,Immunohistochemistry ,In vitro ,Cell biology ,Disease Models, Animal ,Cell Transformation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,biology.protein ,Epidermis ,Stem cell ,Carcinogenesis ,Hair Follicle ,Biomarkers ,Research Article - Abstract
Although hair bulge stem cells are not the cancer cells-of-origin, they contribute to two-stage DMBA/TPA skin carcinogenesis in an α3β1-dependent manner., Epidermal-specific deletion of integrin α3β1 almost completely prevents the formation of papillomas during 7,12-Dimethylbenz[a]anthracene/12-O-tetradecanoylphorbol-13-acetate (DMBA/TPA) two-stage skin carcinogenesis. This dramatic decrease in tumorigenesis was thought to be due to an egress and premature differentiation of α3β1-depleted hair bulge (HB) stem cells (SCs), previously considered to be the cancer cells-of-origin in the DMBA/TPA model. Using a reporter mouse line with inducible deletion of α3β1 in HBs, we show that HB SCs remain confined to their niche regardless of the presence of α3β1 and are largely absent from skin tumors. However, tumor formation was significantly decreased in mice deficient for α3β1 in HB SCs. RNA sequencing of HB SCs isolated from short-term DMBA/TPA–treated skin showed α3β1-dependent expression of the matricellular protein connective tissue growth factor (CCN2), which was confirmed in vitro, where CCN2 promoted colony formation and 3D growth of transformed keratinocytes. Together, these findings show that HBs contribute to skin tumorigenesis in an α3β1-dependent manner and suggest a role of HB SCs in creating a permissive environment for tumor growth through the modulation of CCN2 secretion.
- Published
- 2020
28. Simultaneous measurement of contraction, voltage and calcium in HIPSC-CMS for the detection of inotropic effects under blinded conditions
- Author
-
Chris Denning, Samantha Turner, Eric I. Rossman, Christine L. Mummery, Xiaoping Xu, Anthony Bahinski, Berend J. van Meer, Ana Krotenberg Garcia, Leon G.J. Tertoolen, Tessa de Korte, Peter Clements, Marijn Vlaming, and Stefan R. Braam
- Subjects
Pharmacology ,Inotrope ,medicine.medical_specialty ,Contraction (grammar) ,Chemistry ,Internal medicine ,Hipsc cms ,medicine ,Cardiology ,chemistry.chemical_element ,Calcium ,Toxicology - Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.