31 results on '"Tian TV"'
Search Results
2. Carm1-arginine methylation of the transcription factor C/EBPα regulates transdifferentiation velocity
- Author
-
Torcal Garcia, Guillem, Kowenz-Leutz, Elisabeth, TIAN, TIAN, Klonizakis, Antonis, Lerner, Jonathan, de Andres-Aguayo, Luisa, Vila-Casadesús, Maria, Peiró, Sandra, Institut Català de la Salut, [Torcal Garcia G, Klonizakis A, De Andres-Aguayo L] Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain. Universitat Pompeu Fabra (UPF), Barcelona, Spain. [Kowenz-Leutz E] Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany. [Tian TV, Vila Casadesus M] Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain. Universitat Pompeu Fabra (UPF), Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Lerner J] Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States. [Peiro S] Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Cell Physiological Phenomena::Cell Differentiation [PHENOMENA AND PROCESSES] ,Cell Physiological Phenomena::Cell Transdifferentiation [PHENOMENA AND PROCESSES] ,Cèl·lules B ,fenómenos químicos::fenómenos bioquímicos::alquilación::metilación [FENÓMENOS Y PROCESOS] ,Chemical Phenomena::Biochemical Phenomena::Alkylation::Methylation [PHENOMENA AND PROCESSES] ,Diferenciació cel·lular ,Ratolins ,fenómenos fisiológicos celulares::transdiferenciación celular [FENÓMENOS Y PROCESOS] ,Metilació ,Eukaryota::Animals::Chordata::Vertebrates::Mammals::Eutheria::Rodentia::Muridae::Murinae::Mice [ORGANISMS] ,Eukaryota::animales::Chordata::vertebrados::mamíferos::Eutheria::Rodentia::Muridae::Murinae::ratones [ORGANISMOS] ,fenómenos fisiológicos celulares::diferenciación celular [FENÓMENOS Y PROCESOS] - Abstract
Developmental biology; Gene regulation; Transcription factor Biologia del desenvolupament; Regulació gènica; Factor de transcripció Biología del desarrollo; Regulación génica; Factor de transcripción Here, we describe how the speed of C/EBPα-induced B cell to macrophage transdifferentiation (BMT) can be regulated, using both mouse and human models. The identification of a mutant of C/EBPα (C/EBPαR35A) that greatly accelerates BMT helped to illuminate the mechanism. Thus, incoming C/EBPα binds to PU.1, an obligate partner expressed in B cells, leading to the release of PU.1 from B cell enhancers, chromatin closing and silencing of the B cell program. Released PU.1 redistributes to macrophage enhancers newly occupied by C/EBPα, causing chromatin opening and activation of macrophage genes. All these steps are accelerated by C/EBPαR35A, initiated by its increased affinity for PU.1. Wild-type C/EBPα is methylated by Carm1 at arginine 35 and the enzyme’s perturbations modulate BMT velocity as predicted from the observations with the mutant. Increasing the proportion of unmethylated C/EBPα in granulocyte/macrophage progenitors by inhibiting Carm1 biases the cell’s differentiation toward macrophages, suggesting that cell fate decision velocity and lineage directionality are closely linked processes. TG was supported by the Center for Genomic Regulation, Barcelona, the Spanish Ministry of Economy, Industry and Competitiveness, (Plan Estatal PID2019-109354GB-100), AGAUR (SGR 006713) and the 4D-Genome European Research Council Synergy grant. KSZ was supported by the NIH grant R01GM36477. We have used ChatGPT to improve parts of the text.
- Published
- 2023
3. Human metastatic cholangiocarcinoma patient-derived xenografts and tumoroids for preclinical drug evaluation
- Author
-
Queralt Serra-Camprubí, Helena Verdaguer, Winona Oliveros, Núria Lupión-Garcia, Alba Llop-Guevara, Cristina Molina, Maria Vila-Casadesús, Anthony Turpin, Cindy Neuzillet, Joan Frigola, Jessica Querol, Mariana Yáñez-Bartolomé, Florian Castet, Carles Fabregat-Franco, Carmen Escudero-Iriarte, Marta Escorihuela, Enrique J. Arenas, Cristina Bernadó-Morales, Noemí Haro, Francis J. Giles, Óscar J. Pozo, Josep M. Miquel, Paolo G. Nuciforo, Ana Vivancos, Marta Melé, Violeta Serra, Joaquín Arribas, Josep Tabernero, Sandra Peiró, Teresa Macarulla, Tian V. Tian, Institut Català de la Salut, [Serra-Camprubí Q, Lupión-Garcia N, Llop-Guevara A, Molina C, Querol J, Yáñez-Bartolomé M, Escudero-Iriarte C, Escorihuela M, Arenas EJ, Bernadó-Morales C, Miquel JM, Nuciforo PG, Serra V, Peiró S, Tian TV] Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Verdaguer H, Castet F, Fabregat-Franco C, Tabernero J, Macarulla T] Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Gastrointestinal and Endocrine Tumor Unit, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Vall d’Hebron Hospital Universitari, Barcelona, Spain. [Oliveros W] Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Spain. [Vila-Casadesús M, Vivancos A] Cancer Genomics Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Frigola J] Clinical Research Program, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Arribas J] Preclinical and Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Centro de Investigacion Biomédica en Red de Cáncer, Monforte de Lemos, Madrid, Spain. Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain. Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain. Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain, Vall d'Hebron Barcelona Hospital Campus, and Barcelona Supercomputing Center
- Subjects
Informàtica::Aplicacions de la informàtica::Bioinformàtica [Àrees temàtiques de la UPC] ,neoplasias::neoplasias por tipo histológico::neoplasias glandulares y epiteliales::carcinoma::adenocarcinoma::colangiocarcinoma [ENFERMEDADES] ,Cancer Research ,Neoplasms::Neoplasms by Histologic Type::Neoplasms, Glandular and Epithelial::Carcinoma::Adenocarcinoma::Cholangiocarcinoma [DISEASES] ,Metastatic Cholangiocarcinoma ,Conductes biliars - Càncer - Tractament ,Conductes biliars -- Tumors ,Drug response ,neoplasias::neoplasias por localización::neoplasias del sistema digestivo::neoplasias del tracto biliar::neoplasias de los conductos biliares [ENFERMEDADES] ,Investigative Techniques::Drug Development::Drug Evaluation, Preclinical [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,técnicas de investigación::desarrollo de medicamentos::evaluación preclínica de medicamentos [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Neoplasms::Neoplasms by Site::Digestive System Neoplasms::Biliary Tract Neoplasms::Bile Duct Neoplasms [DISEASES] ,Oncology ,Statistical analysis ,Simulació per ordinador ,Medicaments - Assaigs clínics ,Pathogenic mutations ,Càncer ,Genètica ,Cancer - Abstract
Cholangiocarcinoma (CCA) is usually diagnosed at advanced stages, with limited therapeutic options. Preclinical models focused on unresectable metastatic CCA are necessary to develop rational treatments. Pathogenic mutations in IDH1/2, ARID1A/B, BAP1, and BRCA1/2 have been identified in 30\\%–50\\% of patients with CCA. Several types of tumor cells harboring these mutations exhibit homologous recombination deficiency (HRD) phenotype with enhanced sensitivity to PARP inhibitors (PARPi). However, PARPi treatment has not yet been tested for effectiveness in patient-derived models of advanced CCA.We have established a collection of patient-derived xenografts from patients with unresectable metastatic CCA (CCA\_PDX). The CCA\_PDXs were characterized at both histopathologic and genomic levels. We optimized a protocol to generate CCA tumoroids from CCA\_PDXs. We tested the effects of PARPis in both CCA tumoroids and CCA\_PDXs. Finally, we used the RAD51 assay to evaluate the HRD status of CCA tissues.This collection of CCA\_PDXs recapitulates the histopathologic and molecular features of their original tumors. PARPi treatments inhibited the growth of CCA tumoroids and CCA\_PDXs with pathogenic mutations of BRCA2, but not those with mutations of IDH1, ARID1A, or BAP1. In line with these findings, only CCA\_PDX and CCA patient biopsy samples with mutations of BRCA2 showed RAD51 scores compatible with HRD.Our results suggest that patients with advanced CCA with pathogenic mutations of BRCA2, but not those with mutations of IDH1, ARID1A, or BAP1, are likely to benefit from PARPi therapy. This collection of CCA\_PDXs provides new opportunities for evaluating drug response and prioritizing clinical trials. The authors would like to thank the patients and their families for their support. This work was supported by grants from the Fundaci o Marat o TV3 awarded to T. Macarulla, M. Mel e, and S. Peir o; BeiGene research grant awarded toT. Macarulla and S. Peir o; AECC (INVES20036TIAN), Ram on y Cajal investigator program (RYC2020-029098-I), Proyecto de IþDþi (PID2019-108008RJ-I00), and FERO Foundation grant awarded to T.V. Tian; Proyecto de Investigaci on en Salud from the Instituto de Salud Carlos III (ISCIII) (PI20/00898) awarded to T. Macarulla; FIS/FEDER from the Instituto de Salud Carlos III (ISCIII) (PI12/01250; CP08/00223; PI16/00253 and CB16/12/00449) awarded to S. Peir o; and Ram on y Cajal investigator program (RYC-2017-22249) awarded to M. Mel e. Q. Serra-Camprubí is a recipient of the Ph.D. fellowship from La Caixa Foundation (LCF/PR/PR12/51070001). A. LlopGuevara was supported by the AECC (INVES20095LLOP) and V. Serra by the ISCIII (CPII19/00033). E.J. Arenas was funded by the AECC (POSTD211413AREN).J. Arribas is funded by the Instituto de Salud Carlos III (AC15/00062, CB16/12/00449, and PI22/00001). This publication is based upon the work of COST Action CA18122, European Cholangiocarcinoma Network, supported by the COST (European Cooperation in Science and Technology, www.cost.eu), a funding agency for research and innovation networks. The authors would like to thank Dr. V.A. Raker for manuscript editing and Drs. N. Herranz and J. Mateo for scientific discussions. The authors acknowledge the infrastructure and support of the FERO Foundation, La Caixa Foundation, and the Cellex Foundation. Peer Reviewed "Article signat per 31 autors/es: Queralt Serra-Camprubí; Helena Verdaguer; Winona Oliveros; Núria Lupión-Garcia; Núria Lupión-Garcia;Alba Llop-Guevara; Cristina Molina; Maria Vila-Casadesús; Anthony Turpin; Cindy Neuzillet; Joan Frigola; Jessica Querol; Mariana Yáñez-Bartolomé; Florian Castet; Carles Fabregat-Franco; Carmen Escudero-Iriarte; Marta Escorihuela; Enrique J. Arenas; Cristina Bernadó-Morales; Noemí Haro; Francis J. Giles; Óscar J. Pozo; Josep M. Miquel ; Paolo G. Nuciforo; Ana Vivancos; Marta Melé; Violeta Serra ; Joaquín Arribas; Josep Tabernero; Sandra Peiró; Teresa Macarulla; Tian V. Tian"
- Published
- 2022
4. The trophectoderm acts as a niche for the inner cell mass through C/EBPα-regulated IL-6 signaling
- Author
-
Marcos Plana-Carmona, Gregoire Stik, Romain Bulteau, Carolina Segura-Morales, Noelia Alcázar, Chris D.R. Wyatt, Antonios Klonizakis, Luisa de Andrés-Aguayo, Maxime Gasnier, Tian V. Tian, Guillem Torcal Garcia, Maria Vila-Casadesús, Nicolas Plachta, Manuel Serrano, Mirko Francesconi, Thomas Graf, Institut Català de la Salut, [Plana-Carmona M, Stik G, Segura-Morales C] Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain. Universitat Pompeu Fabra (UPF), Barcelona, Spain. [Bulteau R] Laboratoire de Biologie et Modélisation de la Cellule, Université de Lyon, Lyon, France. [Alcázar N] Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Wyatt CDR] Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain. Universitat Pompeu Fabra (UPF), Barcelona, Spain. Department of Genetics, Evolution & Environment, University College London, London, UK. [Tian TV] Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain. Universitat Pompeu Fabra (UPF), Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Vila-Casadesús M] Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Embryology ,factores biológicos::péptidos y proteínas de señalización intercelular::citocinas::interleucinas::interleucina-6 [COMPUESTOS QUÍMICOS Y DROGAS] ,INNER CELL MASS ,Embryonic Development ,Pre-implantation embryo development ,Biochemistry ,Morula ,Interleucina-6 ,Genetics ,CCAAT-Enhancer-Binding Protein-alpha ,TRANSDIFFERENTIATION ,Gene Regulation ,Physiological Phenomena::Growth and Development::Morphogenesis::Embryonic and Fetal Development::Embryonic Development [PHENOMENA AND PROCESSES] ,Fisiologia cel·lular ,Transdifferentiation ,C/EBP transcription factor ,Embriologia ,Interleukin-6 ,somatic cell reprogramming ,Cell Biology ,Gene regulation ,Blastocyst ,pre-implantation embryo development ,Biological Factors::Intercellular Signaling Peptides and Proteins::Cytokines::Interleukins::Interleukin-6 [CHEMICALS AND DRUGS] ,estructuras embrionarias::mórula [ANATOMÍA] ,Trophectoderm ,IL-6 signaling ,Inner cell mass ,Embryonic Structures::Morula [ANATOMY] ,fenómenos fisiológicos::crecimiento y desarrollo::morfogénesis::desarrollo embrionario y fetal::desarrollo embrionario [FENÓMENOS Y PROCESOS] ,Somatic cell reprogramming ,Developmental Biology - Abstract
Gene regulation; Somatic cell reprogramming; Trophectoderm Regulación de genes; Reprogramación de células somáticas; Trofoectodermo Regulació de gens; Reprogramació de cèl·lules somàtiques; Trofectoderma IL-6 has been shown to be required for somatic cell reprogramming into induced pluripotent stem cells (iPSCs). However, how Il6 expression is regulated and whether it plays a role during embryo development remains unknown. Here, we describe that IL-6 is necessary for C/EBPα-enhanced reprogramming of B cells into iPSCs but not for B cell to macrophage transdifferentiation. C/EBPα overexpression activates both Il6 and Il6ra genes in B cells and in PSCs. In embryo development, Cebpa is enriched in the trophectoderm of blastocysts together with Il6, while Il6ra is mostly expressed in the inner cell mass (ICM). In addition, Il6 expression in blastocysts requires Cebpa. Blastocysts secrete IL-6 and neutralization of the cytokine delays the morula to blastocyst transition. The observed requirement of C/EBPα-regulated IL-6 signaling for pluripotency during somatic cell reprogramming thus recapitulates a physiologic mechanism in which the trophectoderm acts as niche for the ICM through the secretion of IL-6. We thank C. Berenguer for help with B cell reprogramming and bone marrow collection; S. Nakagawa and B. Pernaute for advice on pre-implantation embryo culture and manipulation, and Kyle M. Loh for his valuable discussions; the flow cytometry and microscopy units of UPF-CRG for technical assistance; the CRG genomics core facility for sequencing and Graf laboratory members for critical discussions. Work in the laboratory of T.G. was supported by the Spanish Ministry of Economy, Industry and Competitiveness (Plan Estatal PID2019-109354GB-I00), the CRG, AGAUR (SGR 726), and a European Research Council Synergy grant (4D-Genome). M.P.-C. was supported by an FPI fellowship (BES-2016-076900). Work in the laboratory of M.S. was funded by the IRB and by grants from the Spanish Ministry of Economy co-funded by the European Regional Development Fund (SAF2017-82613-R), ERC (ERC-2014-AdG/669622), la Caixa Foundation, and Secretaria d’Universitats i Recerca del Departament d’Empresa i Coneixement of Catalonia (Grup de Recerca consolidat 2017 SGR 282).
- Published
- 2022
5. Identification of enhancer-promoter contacts in embryoid bodies by quantitative chromosome conformation capture (4C)
- Author
-
Enrique Vidal, Gregoire Stik, Thomas Graf, Tian V. Tian, Institut Català de la Salut, [Tian TV] Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain. Universitat Pompeu Fabra, Barcelona, Spain. Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Vidal E, Graf T, Stik G] Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain. Universitat Pompeu Fabra, Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Pluripotent Stem Cells ,células::células madre::células madre pluripotentes::células madre embrionarias::cuerpos embrioides [ANATOMÍA] ,General Chemical Engineering ,Computational biology ,Embryoid body ,Cell fate determination ,Biology ,Polymerase Chain Reaction ,Chromosomes ,General Biochemistry, Genetics and Molecular Biology ,Chromosome conformation capture ,Embrions ,Mice ,Investigative Techniques::Genetic Techniques::Sequence Analysis::High-Throughput Nucleotide Sequencing [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Animals ,Promoter Regions, Genetic ,Induced pluripotent stem cell ,Enhancer ,Gene ,Embryoid Bodies ,Seqüència de nucleòtids ,Embriologia ,General Immunology and Microbiology ,General Neuroscience ,Computational Biology ,High-Throughput Nucleotide Sequencing ,Promoter ,DNA Restriction Enzymes ,Cells::Stem Cells::Pluripotent Stem Cells::Embryonic Stem Cells::Embryoid Bodies [ANATOMY] ,Cromosomes ,Restriction enzyme ,Enhancer Elements, Genetic ,técnicas de investigación::técnicas genéticas::análisis de secuencias::secuenciación de nucleótidos de alto rendimiento [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Genètica - Abstract
Cèl·lules mare embrionàries; Seqüenciació d'alt rendiment Células madre embrionarias; Secuenciación de alto rendimiento Embryonic stem cells; High-throughput sequencing During mammalian development, cell fates are determined through the establishment of regulatory networks that define the specificity, timing, and spatial patterns of gene expression. Embryoid bodies (EBs) derived from pluripotent stem cells have been a popular model to study the differentiation of the main three germ layers and to define regulatory circuits during cell fate specification. Although it is well-known that tissue-specific enhancers play an important role in these networks by interacting with promoters, assigning them to their relevant target genes still remains challenging. To make this possible, quantitative approaches are needed to study enhancer-promoter contacts and their dynamics during development. Here, we adapted a 4C method to define enhancers and their contacts with cognate promoters in the EB differentiation model. The method uses frequently cutting restriction enzymes, sonication, and a nested-ligation-mediated PCR protocol compatible with commercial DNA library preparation kits. Subsequently, the 4C libraries are subjected to high-throughput sequencing and analyzed bioinformatically, allowing detection and quantification of all sequences that have contacts with a chosen promoter. The resulting sequencing data can also be used to gain information about the dynamics of enhancer-promoter contacts during differentiation. The technique described for the EB differentiation model is easy to implement. We would like to thank F. Le Dily, R. Stadhouders and members of the Graf laboratory for their advice and discussions. G.S. was supported by a Marie Sklodowska-Curie fellowship (H2020-MSCA-IF-2016, miRStem), T.V.T by a Juan de la Cierva postdoctoral fellowship (MINECO, FJCI-2014-22946). This work was supported by the European Research Council under the 7th Framework Programme FP7 (ERC Synergy Grant 4D-Genome, grant agreement 609989 to T.G.), the Spanish Ministry of Economy, Industry and Competitiveness (MEIC) to the EMBL partnership, Centro de Excelencia Severo Ochoa 2013-2017 and CERCA Program Generalitat de Catalunya.
- Published
- 2020
6. Promising response to lurbinectedin in NUT carcinoma: a case report and review of emerging therapeutic strategies.
- Author
-
Sánchez Becerra MV, Escudero Iriarte C, Travert C, Tian TV, and Besse B
- Published
- 2024
- Full Text
- View/download PDF
7. Clinical Value of Liquid Biopsy in Patients with FGFR2 Fusion-Positive Cholangiocarcinoma During Targeted Therapy.
- Author
-
González-Medina A, Vila-Casadesús M, Gomez-Rey M, Fabregat-Franco C, Sierra A, Tian TV, Castet F, Castillo G, Matito J, Martinez P, Miquel JM, Nuciforo P, Pérez-López R, Macarulla T, and Vivancos A
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Liquid Biopsy methods, Retrospective Studies, Oncogene Proteins, Fusion genetics, Molecular Targeted Therapy methods, Circulating Tumor DNA genetics, Circulating Tumor DNA blood, High-Throughput Nucleotide Sequencing methods, Adult, Prognosis, Protein Kinase Inhibitors therapeutic use, Cholangiocarcinoma drug therapy, Cholangiocarcinoma genetics, Cholangiocarcinoma pathology, Cholangiocarcinoma blood, Cholangiocarcinoma diagnosis, Receptor, Fibroblast Growth Factor, Type 2 genetics, Biomarkers, Tumor genetics, Biomarkers, Tumor blood, Bile Duct Neoplasms drug therapy, Bile Duct Neoplasms genetics, Bile Duct Neoplasms pathology, Bile Duct Neoplasms blood, Bile Duct Neoplasms mortality
- Abstract
Purpose: FGFR2 fusions occur in 10% to 15% of patients with intrahepatic cholangiocarcinoma (iCCA), potentially benefiting from FGFR inhibitors (FGFRi). We aimed to assess the feasibility of detecting FGFR2 fusions in plasma and explore plasma biomarkers for managing FGFRi treatment., Experimental Design: We conducted a retrospective study in 18 patients with iCCA and known FGFR2 fusions previously identified in tissue samples from prior FGFRi treatment. Both tissue and synchronous plasma samples were analyzed using a custom hybrid capture gene panel with next-generation sequencing (VHIO-iCCA panel) and validated against commercial vendor results. Longitudinal plasma analysis during FGFRi was performed. Subsequently, we explored the correlation between plasma biomarkers, liver enzymes, tumor volume, and clinical outcomes., Results: Sixteen patients (88.9%) were positive for FGFR2 fusion events in plasma. Remarkably, the analysis of plasma suggests that lower levels of ctDNA are linked to clinical benefits from targeted therapy and result in improved progression-free survival and overall survival. Higher concentrations of cell-free DNA before FGFRi treatment were linked to worse overall survival, correlating with impaired liver function and indicating compromised cell-free DNA removal by the liver. Additionally, increased ctDNA or the emergence of resistance mutations allowed earlier detection of disease progression compared with standard radiologic imaging methods., Conclusions: VHIO-iCCA demonstrated accurate detection of FGFR2 fusions in plasma. The integration of information from various plasma biomarkers holds the potential to predict clinical outcomes and identify treatment failure prior to radiologic progression, offering valuable guidance for the clinical management of patients with iCCA., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
8. LOXL2-mediated chromatin compaction is required to maintain the oncogenic properties of triple-negative breast cancer cells.
- Author
-
Serra-Bardenys G, Blanco E, Escudero-Iriarte C, Serra-Camprubí Q, Querol J, Pascual-Reguant L, Morancho B, Escorihuela M, Tissera NS, Sabé A, Martín L, Segura-Bayona S, Verde G, Aiese Cigliano R, Millanes-Romero A, Jerónimo C, Cebrià-Costa JP, Nuciforo P, Simonetti S, Viaplana C, Dienstmann R, Oliveira M, Peg V, Stracker TH, Arribas J, Canals F, Villanueva J, Di Croce L, García de Herreros A, Tian TV, and Peiró S
- Subjects
- Female, Humans, Cell Line, Tumor, Chromatin metabolism, Chromatin genetics, DNA Helicases genetics, DNA Helicases metabolism, Gene Expression Regulation, Neoplastic, Amino Acid Oxidoreductases genetics, Amino Acid Oxidoreductases metabolism, Heterochromatin metabolism, Heterochromatin genetics, Histones metabolism, Histones genetics, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Triple Negative Breast Neoplasms metabolism
- Abstract
Oxidation of histone H3 at lysine 4 (H3K4ox) is catalyzed by lysyl oxidase homolog 2 (LOXL2). This histone modification is enriched in heterochromatin in triple-negative breast cancer (TNBC) cells and has been linked to the maintenance of compacted chromatin. However, the molecular mechanism underlying this maintenance is still unknown. Here, we show that LOXL2 interacts with RuvB-Like 1 (RUVBL1), RuvB-Like 2 (RUVBL2), Actin-like protein 6A (ACTL6A), and DNA methyltransferase 1associated protein 1 (DMAP1), a complex involved in the incorporation of the histone variant H2A.Z. Our experiments indicate that this interaction and the active form of RUVBL2 are required to maintain LOXL2-dependent chromatin compaction. Genome-wide experiments showed that H2A.Z, RUVBL2, and H3K4ox colocalize in heterochromatin regions. In the absence of LOXL2 or RUVBL2, global levels of the heterochromatin histone mark H3K9me3 were strongly reduced, and the ATAC-seq signal in the H3K9me3 regions was increased. Finally, we observed that the interplay between these series of events is required to maintain H3K4ox-enriched heterochromatin regions, which in turn is key for maintaining the oncogenic properties of the TNBC cell line tested (MDA-MB-231)., (© 2024 Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
9. SRC inhibition enables formation of a growth suppressive MAGI1-PP2A complex in isocitrate dehydrogenase-mutant cholangiocarcinoma.
- Author
-
Luk IS, Bridgwater CM, Yu A, Boila LD, Yáñez-Bartolomé M, Lampano AE, Hulahan TS, Boukhali M, Kathiresan M, Macarulla T, Kenerson HL, Yamamoto N, Sokolov D, Engstrom IA, Sullivan LB, Lampe PD, Cooper JA, Yeung RS, Tian TV, Haas W, Saha SK, and Kugel S
- Subjects
- Animals, Humans, Mice, Bile Duct Neoplasms pathology, Bile Duct Neoplasms metabolism, Bile Duct Neoplasms genetics, Bile Duct Neoplasms drug therapy, Cell Adhesion Molecules metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Phosphorylation drug effects, Ribosomal Protein S6 Kinases, 70-kDa metabolism, Signal Transduction drug effects, Guanylate Kinases genetics, Guanylate Kinases metabolism, Protein Phosphatase 2 genetics, Protein Phosphatase 2 metabolism, Adaptor Proteins, Signal Transducing metabolism, Cholangiocarcinoma drug therapy, Cholangiocarcinoma pathology, Cholangiocarcinoma metabolism, Cholangiocarcinoma genetics, Dasatinib pharmacology, Isocitrate Dehydrogenase metabolism, Isocitrate Dehydrogenase genetics, Mutation genetics, src-Family Kinases metabolism, src-Family Kinases antagonists & inhibitors
- Abstract
Intrahepatic cholangiocarcinoma (ICC) is an aggressive bile duct malignancy that frequently exhibits isocitrate dehydrogenase ( IDH1/IDH2 ) mutations. Mutant IDH (IDHm) ICC is dependent on SRC kinase for growth and survival and is hypersensitive to inhibition by dasatinib, but the molecular mechanism underlying this sensitivity is unclear. We found that dasatinib reduced p70 S6 kinase (S6K) and ribosomal protein S6 (S6), leading to substantial reductions in cell size and de novo protein synthesis. Using an unbiased phosphoproteomic screen, we identified membrane-associated guanylate kinase, WW, and PDZ domain containing 1 (MAGI1) as an SRC substrate in IDHm ICC. Biochemical and functional assays further showed that SRC inhibits a latent tumor-suppressing function of the MAGI1-protein phosphatase 2A (PP2A) complex to activate S6K/S6 signaling in IDHm ICC. Inhibiting SRC led to activation and increased access of PP2A to dephosphorylate S6K, resulting in cell death. Evidence from patient tissue and cell line models revealed that both intrinsic and extrinsic resistance to dasatinib is due to increased phospho-S6 (pS6). To block pS6, we paired dasatinib with the S6K/AKT inhibitor M2698, which led to a marked reduction in pS6 in IDHm ICC cell lines and patient-derived organoids in vitro and substantial growth inhibition in ICC patient-derived xenografts in vivo. Together, these results elucidated the mechanism of action of dasatinib in IDHm ICC, revealed a signaling complex regulating S6K phosphorylation independent of mTOR, suggested markers for dasatinib sensitivity, and described a combination therapy for IDHm ICC that may be actionable in the clinic.
- Published
- 2024
- Full Text
- View/download PDF
10. Generation of Metastatic Cholangiocarcinoma Patient-Derived Xenograft Models.
- Author
-
Yáñez-Bartolomé M, Serra-Camprubí Q, Arenas EJ, Escorihuela M, Castet F, Fabregat-Franco C, Querol J, Arribas J, Peiró S, Macarulla T, and Tian TV
- Subjects
- Humans, Animals, Mice, Disease Models, Animal, Cholangiocarcinoma pathology, Cholangiocarcinoma genetics, Bile Duct Neoplasms pathology, Bile Duct Neoplasms genetics, Xenograft Model Antitumor Assays methods
- Abstract
Cholangiocarcinoma (CCA) poses a substantial clinical hurdle as it is often detected at advanced metastatic stages with limited therapeutic options. To enhance our understanding of advanced CCA, it is imperative to establish preclinical models that faithfully recapitulate the disease's characteristics. Patient-derived xenograft (PDX) models have emerged as a valuable approach in cancer research, offering an avenue to reproduce and study the genomic, histologic, and molecular features of the original human tumors. By faithfully preserving the heterogeneity, microenvironmental interactions, and drug responses observed in human tumors, PDX models serve as highly relevant and predictive preclinical tools. Here, we present a comprehensive protocol that outlines the step-by-step process of generating and maintaining PDX models using biopsy samples from patients with advanced metastatic CCA. The protocol encompasses crucial aspects such as tissue processing, xenograft transplantation, and subsequent monitoring of the PDX models. By employing this protocol, we aim to establish a robust collection of PDX models that accurately reflect the genomic landscape, histologic diversity, and therapeutic responses observed in advanced CCA, thereby enabling improved translational research, drug development, and personalized treatment strategies for patients facing this challenging disease., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
11. Interactions between BRD4S, LOXL2, and MED1 drive cell cycle transcription in triple-negative breast cancer.
- Author
-
Pascual-Reguant L, Serra-Camprubí Q, Datta D, Cianferoni D, Kourtis S, Gañez-Zapater A, Cannatá C, Espinar L, Querol J, García-López L, Musa-Afaneh S, Guirola M, Gkanogiannis A, Miró Canturri A, Guzman M, Rodríguez O, Herencia-Ropero A, Arribas J, Serra V, Serrano L, Tian TV, Peiró S, and Sdelci S
- Subjects
- Humans, Amino Acid Oxidoreductases genetics, Amino Acid Oxidoreductases metabolism, Bromodomain Containing Proteins, Cell Cycle, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Proliferation genetics, Gene Expression Regulation, Neoplastic, Mediator Complex Subunit 1 genetics, Mediator Complex Subunit 1 metabolism, Nuclear Proteins genetics, Animals, Transcription Factors metabolism, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms metabolism
- Abstract
Triple-negative breast cancer (TNBC) often develops resistance to single-agent treatment, which can be circumvented using targeted combinatorial approaches. Here, we demonstrate that the simultaneous inhibition of LOXL2 and BRD4 synergistically limits TNBC proliferation in vitro and in vivo. Mechanistically, LOXL2 interacts in the nucleus with the short isoform of BRD4 (BRD4S), MED1, and the cell cycle transcriptional regulator B-MyB. These interactions sustain the formation of BRD4 and MED1 nuclear transcriptional foci and control cell cycle progression at the gene expression level. The pharmacological co-inhibition of LOXL2 and BRD4 reduces BRD4 nuclear foci, BRD4-MED1 colocalization, and the transcription of cell cycle genes, thus suppressing TNBC cell proliferation. Targeting the interaction between BRD4S and LOXL2 could be a starting point for the development of new anticancer strategies for the treatment of TNBC., (© 2023 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
12. Fascin-1 expression is associated with neuroendocrine prostate cancer and directly suppressed by androgen receptor.
- Author
-
Turpin A, Delliaux C, Parent P, Chevalier H, Escudero-Iriarte C, Bonardi F, Vanpouille N, Flourens A, Querol J, Carnot A, Leroy X, Herranz N, Lanel T, Villers A, Olivier J, Touzet H, de Launoit Y, Tian TV, and Duterque-Coquillaud M
- Subjects
- Humans, Male, Androgen Antagonists therapeutic use, Androgens, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Microfilament Proteins genetics, Microfilament Proteins metabolism, Neuroendocrine Tumors genetics, Neuroendocrine Tumors pathology, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Receptors, Androgen genetics, Receptors, Androgen metabolism
- Abstract
Background: Neuroendocrine prostate cancer (NEPC) is an aggressive form of prostate cancer, arising from resistance to androgen-deprivation therapies. However, the molecular mechanisms associated with NEPC development and invasiveness are still poorly understood. Here we investigated the expression and functional significance of Fascin-1 (FSCN1), a pro-metastasis actin-bundling protein associated with poor prognosis of several cancers, in neuroendocrine differentiation of prostate cancer., Methods: Differential expression analyses using Genome Expression Omnibus (GEO) database, clinical samples and cell lines were performed. Androgen or antagonist's cellular treatments and knockdown experiments were used to detect changes in cell morphology, molecular markers, migration properties and in vivo tumour growth. Chromatin immunoprecipitation-sequencing (ChIP-Seq) data and ChIP assays were analysed to decipher androgen receptor (AR) binding., Results: We demonstrated that FSCN1 is upregulated during neuroendocrine differentiation of prostate cancer in vitro, leading to phenotypic changes and NEPC marker expression. In human prostate cancer samples, FSCN1 expression is restricted to NEPC tumours. We showed that the androgen-activated AR downregulates FSCN1 expression and works as a transcriptional repressor to directly suppress FSCN1 expression. AR antagonists alleviate this repression. In addition, FSCN1 silencing further impairs in vivo tumour growth., Conclusion: Collectively, our findings identify FSCN1 as an AR-repressed gene. Particularly, it is involved in NEPC aggressiveness. Our results provide the rationale for the future clinical development of FSCN1 inhibitors in NEPC patients., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
13. The potential of patient-derived organoids in precision medicine of biliary tract cancer.
- Author
-
Yáñez-Bartolomé M, Macarulla T, and Tian TV
- Subjects
- Humans, Organoids, Precision Medicine, Biliary Tract Neoplasms drug therapy, Biliary Tract Neoplasms genetics
- Abstract
Chemotherapy resistance in biliary tract cancer (BTC) presents a major clinical hurdle. Ren et al.
1 developed and characterized an extensive collection of BTC patient-derived organoid (PDO) models, enabling advanced investigation of chemotherapy response prediction., Competing Interests: Declaration of interests T.M. reports scientific consultancy role for Ability Pharmaceuticals SL, AstraZeneca, Basilea Pharma, Baxter, BioLineRX Ltd, Celgene, Eisai, Incyte, Ipsen Bioscience Inc, speaker’s fee for Janssen and Lilly, and research funding for MSD, Novocure, QED Therapeutics, Roche Farma, Sanofi-Aventis, Servier, and Zymeworks. T.V.T. reports grants from Loxo Oncology at Lilly, Pharmaxis, Alentis, Incyte, and nonfinancial support from Servier., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
14. Clinical and genomic characterisation of early-onset pancreatic cancer.
- Author
-
Castet F, Fabregat-Franco C, Castillo G, Navarro V, Sierra A, Acosta DA, López-Valbuena D, Dienstmann R, Tabernero J, Vivancos A, Tian TV, and Macarulla T
- Subjects
- Humans, Retrospective Studies, Genomics, Precision Medicine adverse effects, Pancreatic Neoplasms epidemiology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms diagnosis
- Abstract
Background: The incidence of early-onset pancreatic cancer (EOPC) has risen dramatically in recent years. We aimed to characterise the clinical and genomic features of EOPC and evaluate their therapeutic implications., Methods: We performed a comparative, single-centre, retrospective analysis of clinical, germline, and genomic features in EOPC (≤50 years) patients and compared them with a control group of average-onset pancreatic cancer patients (AOPC, ≥70 years). Key molecular findings were compared with an external, publicly available cohort., Results: We reviewed 336 patients who met all inclusion criteria (EOPC N = 139, AOPC N = 197). EOPC was associated with smoking status, lower prevalence of diabetes, better performance status, higher CA19.9 levels, and higher albumin levels at diagnosis. After adjustment for baseline covariates, we observed no differences in overall survival (OS). Age was associated with an increase in the incidence of KRAS
MUT both in our cohort and the validation cohort. EOPC were enriched in potentially actionable alterations according to ESCAT tiers I-IIIA when compared with AOPC in discovery and validation cohorts (19% versus 14% and 14% versus 8%, respectively). In the first-line metastatic setting, EOPC had a longer progression-free survival (hazard ratio [HR] 0.61, 95% confidence interval (CI) 0.43-0.87) and OS (HR 0.65, 95% CI 0.45-0.95), although there were no differences in response rate. After adjusting for the number of treatment lines, EOPC patients who did receive targeted therapies exhibited longer OS compared with EOPC who did not (HR 0.34, 95% CI 0.12-0.93)., Conclusions: EOPC patients have improved outcomes in the metastatic setting when compared to AOPC and are enriched for targetable alterations that open opportunities for precision oncology-based approaches., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Rodrigo Dienstmann reports an advisory role for Roche, Foundation Medicine, received a speaker’s fee from Roche, Ipsen, Amgen, Servier, Sanofi, Libbs, Merck Sharp & Dohme, Lilly, AstraZeneca, Janssen, Takeda, GlaxoSmithKline, Gilead, and research grants from Merck, Novartis, Daiichi-Sankyo, GlaxoSmithKline, and AstraZeneca. Josep Tabernero reports personal financial interest in the form of scientific consultancy role for Array Biopharma, AstraZeneca, Bayer, Boehringer Ingelheim, Cardiff Oncology, Chugai, Daiichi-Sankyo, F. Hoffmann-La Roche Ltd, Genentech Inc, HalioDX SAS, Hutchison MediPharma International, Ikena Oncology, Inspirna Inc, IQVIA, Lilly, Menarini, Merck Serono, Merus, MSD, Mirati, Neophore, Novartis, Ona Therapeutics, Orion Biotechnology, Peptomyc, Pfizer, Pierre Fabre, Samsung Bioepis, Sanofi, Scandion Oncology, Scorpion Therapeutics, Seattle Genetics, Servier, Sotio Biotech, Taiho, Tessa Therapeutics, TheraMyc, and Tolremo Therapeutics; Stocks: Oniria Therapeutics and also educational collaboration with Imedex/HMP, Medscape Education, MJH Life Sciences, PeerView Institute for Medical Education and Physicians Education Resource (PER). Ana Vivancos reports advisory boards from Bayer, Bristol Meyers Squibb, Guardant Health, Incyte, Roche. Stocks or Shares: Reveal Genomics. Research grant, institutional, financial interest, preclinical research grant: Incyte and Roche. Tian V. Tian reports grants from Loxo Oncology at Lilly, Pharmaxis, Incyte, and Alentis, and non-financial support from Servier that are not directly associated with the submitted work. Teresa Macarulla reports personal financial interest in the form of scientific consultancy role for Ability Pharmaceuticals SL, AstraZeneca, Basilea Pharma, Baxter, BioLineRX Ltd, Celgene, Eisai, Incyte, Ipsen Bioscience Inc; speaker’s fee for Janssen and Lilly and direct research funding for MSD, Novocure, QED Therapeutics, Roche Farma, Sanofi-Aventis, Servier, Zymeworks. The remaining authors declare no potential conflicts of interest., (Copyright © 2023. Published by Elsevier Ltd.)- Published
- 2023
- Full Text
- View/download PDF
15. Carm1-arginine methylation of the transcription factor C/EBPα regulates transdifferentiation velocity.
- Author
-
Torcal Garcia G, Kowenz-Leutz E, Tian TV, Klonizakis A, Lerner J, De Andres-Aguayo L, Sapozhnikova V, Berenguer C, Carmona MP, Casadesus MV, Bulteau R, Francesconi M, Peiro S, Mertins P, Zaret K, Leutz A, and Graf T
- Subjects
- Animals, Humans, Mice, Cell Differentiation genetics, Chromatin, Methylation, Proto-Oncogene Proteins metabolism, CCAAT-Enhancer-Binding Protein-alpha genetics, CCAAT-Enhancer-Binding Protein-alpha metabolism, Cell Transdifferentiation, Trans-Activators genetics, Trans-Activators metabolism
- Abstract
Here, we describe how the speed of C/EBPα-induced B cell to macrophage transdifferentiation (BMT) can be regulated, using both mouse and human models. The identification of a mutant of C/EBPα (C/EBPα
R35A ) that greatly accelerates BMT helped to illuminate the mechanism. Thus, incoming C/EBPα binds to PU.1, an obligate partner expressed in B cells, leading to the release of PU.1 from B cell enhancers, chromatin closing and silencing of the B cell program. Released PU.1 redistributes to macrophage enhancers newly occupied by C/EBPα, causing chromatin opening and activation of macrophage genes. All these steps are accelerated by C/EBPαR35A , initiated by its increased affinity for PU.1. Wild-type C/EBPα is methylated by Carm1 at arginine 35 and the enzyme's perturbations modulate BMT velocity as predicted from the observations with the mutant. Increasing the proportion of unmethylated C/EBPα in granulocyte/macrophage progenitors by inhibiting Carm1 biases the cell's differentiation toward macrophages, suggesting that cell fate decision velocity and lineage directionality are closely linked processes., Competing Interests: GT, EK, TT, AK, JL, LD, VS, CB, MC, MC, RB, MF, SP, PM, KZ, AL, TG No competing interests declared, (© 2023, Torcal Garcia, Kowenz-Leutz, Tian et al.)- Published
- 2023
- Full Text
- View/download PDF
16. Human Metastatic Cholangiocarcinoma Patient-Derived Xenografts and Tumoroids for Preclinical Drug Evaluation.
- Author
-
Serra-Camprubí Q, Verdaguer H, Oliveros W, Lupión-Garcia N, Llop-Guevara A, Molina C, Vila-Casadesús M, Turpin A, Neuzillet C, Frigola J, Querol J, Yáñez-Bartolomé M, Castet F, Fabregat-Franco C, Escudero-Iriarte C, Escorihuela M, Arenas EJ, Bernadó-Morales C, Haro N, Giles FJ, Pozo ÓJ, Miquel JM, Nuciforo PG, Vivancos A, Melé M, Serra V, Arribas J, Tabernero J, Peiró S, Macarulla T, and Tian TV
- Subjects
- Humans, Drug Evaluation, Preclinical, Heterografts, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Bile Ducts, Intrahepatic, Cholangiocarcinoma drug therapy, Cholangiocarcinoma genetics, Bile Duct Neoplasms drug therapy, Bile Duct Neoplasms genetics
- Abstract
Purpose: Cholangiocarcinoma (CCA) is usually diagnosed at advanced stages, with limited therapeutic options. Preclinical models focused on unresectable metastatic CCA are necessary to develop rational treatments. Pathogenic mutations in IDH1/2, ARID1A/B, BAP1, and BRCA1/2 have been identified in 30%-50% of patients with CCA. Several types of tumor cells harboring these mutations exhibit homologous recombination deficiency (HRD) phenotype with enhanced sensitivity to PARP inhibitors (PARPi). However, PARPi treatment has not yet been tested for effectiveness in patient-derived models of advanced CCA., Experimental Design: We have established a collection of patient-derived xenografts from patients with unresectable metastatic CCA (CCA_PDX). The CCA_PDXs were characterized at both histopathologic and genomic levels. We optimized a protocol to generate CCA tumoroids from CCA_PDXs. We tested the effects of PARPis in both CCA tumoroids and CCA_PDXs. Finally, we used the RAD51 assay to evaluate the HRD status of CCA tissues., Results: This collection of CCA_PDXs recapitulates the histopathologic and molecular features of their original tumors. PARPi treatments inhibited the growth of CCA tumoroids and CCA_PDXs with pathogenic mutations of BRCA2, but not those with mutations of IDH1, ARID1A, or BAP1. In line with these findings, only CCA_PDX and CCA patient biopsy samples with mutations of BRCA2 showed RAD51 scores compatible with HRD., Conclusions: Our results suggest that patients with advanced CCA with pathogenic mutations of BRCA2, but not those with mutations of IDH1, ARID1A, or BAP1, are likely to benefit from PARPi therapy. This collection of CCA_PDXs provides new opportunities for evaluating drug response and prioritizing clinical trials., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
17. pTINCR microprotein promotes epithelial differentiation and suppresses tumor growth through CDC42 SUMOylation and activation.
- Author
-
Boix O, Martinez M, Vidal S, Giménez-Alejandre M, Palenzuela L, Lorenzo-Sanz L, Quevedo L, Moscoso O, Ruiz-Orera J, Ximénez-Embún P, Ciriaco N, Nuciforo P, Stephan-Otto Attolini C, Albà MM, Muñoz J, Tian TV, Varela I, Vivancos A, Ramón Y Cajal S, Muñoz P, Rivas C, and Abad M
- Subjects
- Humans, rho GTP-Binding Proteins metabolism, Ubiquitins metabolism, Neoplasms genetics, Sumoylation, RNA, Long Noncoding genetics
- Abstract
The human transcriptome contains thousands of small open reading frames (sORFs) that encode microproteins whose functions remain largely unexplored. Here, we show that TINCR lncRNA encodes pTINCR, an evolutionary conserved ubiquitin-like protein (UBL) expressed in many epithelia and upregulated upon differentiation and under cellular stress. By gain- and loss-of-function studies, we demonstrate that pTINCR is a key inducer of epithelial differentiation in vitro and in vivo. Interestingly, low expression of TINCR associates with worse prognosis in several epithelial cancers, and pTINCR overexpression reduces malignancy in patient-derived xenografts. At the molecular level, pTINCR binds to SUMO through its SUMO interacting motif (SIM) and to CDC42, a Rho-GTPase critical for actin cytoskeleton remodeling and epithelial differentiation. Moreover, pTINCR increases CDC42 SUMOylation and promotes its activation, triggering a pro-differentiation cascade. Our findings suggest that the microproteome is a source of new regulators of cell identity relevant for cancer., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
18. The trophectoderm acts as a niche for the inner cell mass through C/EBPα-regulated IL-6 signaling.
- Author
-
Plana-Carmona M, Stik G, Bulteau R, Segura-Morales C, Alcázar N, Wyatt CDR, Klonizakis A, de Andrés-Aguayo L, Gasnier M, Tian TV, Torcal Garcia G, Vila-Casadesús M, Plachta N, Serrano M, Francesconi M, and Graf T
- Subjects
- Blastocyst, Embryonic Development, Morula metabolism, CCAAT-Enhancer-Binding Protein-alpha genetics, CCAAT-Enhancer-Binding Protein-alpha metabolism, Interleukin-6 metabolism
- Abstract
IL-6 has been shown to be required for somatic cell reprogramming into induced pluripotent stem cells (iPSCs). However, how Il6 expression is regulated and whether it plays a role during embryo development remains unknown. Here, we describe that IL-6 is necessary for C/EBPα-enhanced reprogramming of B cells into iPSCs but not for B cell to macrophage transdifferentiation. C/EBPα overexpression activates both Il6 and Il6ra genes in B cells and in PSCs. In embryo development, Cebpa is enriched in the trophectoderm of blastocysts together with Il6, while Il6ra is mostly expressed in the inner cell mass (ICM). In addition, Il6 expression in blastocysts requires Cebpa. Blastocysts secrete IL-6 and neutralization of the cytokine delays the morula to blastocyst transition. The observed requirement of C/EBPα-regulated IL-6 signaling for pluripotency during somatic cell reprogramming thus recapitulates a physiologic mechanism in which the trophectoderm acts as niche for the ICM through the secretion of IL-6., Competing Interests: Conflicts of interest The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
19. Evidence for additive and synergistic action of mammalian enhancers during cell fate determination.
- Author
-
Choi J, Lysakovskaia K, Stik G, Demel C, Söding J, Tian TV, Graf T, and Cramer P
- Subjects
- CCAAT-Enhancer-Binding Proteins metabolism, Chromatin metabolism, Enhancer Elements, Genetic, Humans, Promoter Regions, Genetic, THP-1 Cells, CCAAT-Enhancer-Binding Proteins genetics, Cell Differentiation genetics, Histones metabolism, RNA metabolism, Transcription, Genetic
- Abstract
Enhancer activity drives cell differentiation and cell fate determination, but it remains unclear how enhancers cooperate during these processes. Here we investigate enhancer cooperation during transdifferentiation of human leukemia B-cells to macrophages. Putative enhancers are established by binding of the pioneer factor C/EBPα followed by chromatin opening and enhancer RNA (eRNA) synthesis from H3K4-monomethylated regions. Using eRNA synthesis as a proxy for enhancer activity, we find that most putative enhancers cooperate in an additive way to regulate transcription of assigned target genes. However, transcription from 136 target genes depends exponentially on the summed activity of its putative paired enhancers, indicating that these enhancers cooperate synergistically. The target genes are cell type-specific, suggesting that enhancer synergy can contribute to cell fate determination. Enhancer synergy appears to depend on cell type-specific transcription factors, and such interacting enhancers are not predicted from occupancy or accessibility data that are used to detect superenhancers., Competing Interests: JC, KL, GS, CD, JS, TT, TG, PC No competing interests declared, (© 2021, Choi et al.)
- Published
- 2021
- Full Text
- View/download PDF
20. Uncovering Sequence-Specific Transcription Factors Interacting with TET2.
- Author
-
Tian TV and Sardina JL
- Subjects
- Dioxygenases, Humans, DNA Methylation, DNA-Binding Proteins metabolism, Immunoprecipitation methods, Protein Interaction Domains and Motifs, Proto-Oncogene Proteins metabolism, Transcription Factors metabolism
- Abstract
Ten-eleven Translocation (TET) enzymes are methylcytosine dioxygenases that are involved in multiple cellular processes, including cellular differentiation and forced cell fate conversions. However, deciphering the molecular mechanisms underlying epigenetic control exerted by these proteins has been hampered by technical limitations, which prevent the identification of essential partners that work in concert with these enzymes to modulate gene expression. In this chapter, we provide a comprehensive description of cutting-edge methods designed to assess physical interactions between sequence-specific transcription factors and the TET2 enzyme.
- Published
- 2021
- Full Text
- View/download PDF
21. CTCF is dispensable for immune cell transdifferentiation but facilitates an acute inflammatory response.
- Author
-
Stik G, Vidal E, Barrero M, Cuartero S, Vila-Casadesús M, Mendieta-Esteban J, Tian TV, Choi J, Berenguer C, Abad A, Borsari B, le Dily F, Cramer P, Marti-Renom MA, Stadhouders R, and Graf T
- Subjects
- Antigens, Differentiation metabolism, CCCTC-Binding Factor genetics, Cell Line, Tumor, Cell Proliferation physiology, Chromatin physiology, Gene Expression Regulation, Humans, Molecular Conformation, Myelopoiesis genetics, Protein Conformation, B-Lymphocytes physiology, CCCTC-Binding Factor physiology, Macrophages physiology, Myelopoiesis physiology
- Abstract
Three-dimensional organization of the genome is important for transcriptional regulation
1-7 . In mammals, CTCF and the cohesin complex create submegabase structures with elevated internal chromatin contact frequencies, called topologically associating domains (TADs)8-12 . Although TADs can contribute to transcriptional regulation, ablation of TAD organization by disrupting CTCF or the cohesin complex causes modest gene expression changes13-16 . In contrast, CTCF is required for cell cycle regulation17 , embryonic development and formation of various adult cell types18 . To uncouple the role of CTCF in cell-state transitions and cell proliferation, we studied the effect of CTCF depletion during the conversion of human leukemic B cells into macrophages with minimal cell division. CTCF depletion disrupts TAD organization but not cell transdifferentiation. In contrast, CTCF depletion in induced macrophages impairs the full-blown upregulation of inflammatory genes after exposure to endotoxin. Our results demonstrate that CTCF-dependent genome topology is not strictly required for a functional cell-fate conversion but facilitates a rapid and efficient response to an external stimulus.- Published
- 2020
- Full Text
- View/download PDF
22. Identification of Enhancer-Promoter Contacts in Embryoid Bodies by Quantitative Chromosome Conformation Capture (4C).
- Author
-
Tian TV, Vidal E, Graf T, and Stik G
- Subjects
- Animals, Chromosomes, Computational Biology, DNA Restriction Enzymes, Enhancer Elements, Genetic, High-Throughput Nucleotide Sequencing, Mice, Pluripotent Stem Cells cytology, Polymerase Chain Reaction, Promoter Regions, Genetic, Embryoid Bodies cytology
- Abstract
During mammalian development, cell fates are determined through the establishment of regulatory networks that define the specificity, timing, and spatial patterns of gene expression. Embryoid bodies (EBs) derived from pluripotent stem cells have been a popular model to study the differentiation of the main three germ layers and to define regulatory circuits during cell fate specification. Although it is well-known that tissue-specific enhancers play an important role in these networks by interacting with promoters, assigning them to their relevant target genes still remains challenging. To make this possible, quantitative approaches are needed to study enhancer-promoter contacts and their dynamics during development. Here, we adapted a 4C method to define enhancers and their contacts with cognate promoters in the EB differentiation model. The method uses frequently cutting restriction enzymes, sonication, and a nested-ligation-mediated PCR protocol compatible with commercial DNA library preparation kits. Subsequently, the 4C libraries are subjected to high-throughput sequencing and analyzed bioinformatically, allowing detection and quantification of all sequences that have contacts with a chosen promoter. The resulting sequencing data can also be used to gain information about the dynamics of enhancer-promoter contacts during differentiation. The technique described for the EB differentiation model is easy to implement.
- Published
- 2020
- Full Text
- View/download PDF
23. Whsc1 links pluripotency exit with mesendoderm specification.
- Author
-
Tian TV, Di Stefano B, Stik G, Vila-Casadesús M, Sardina JL, Vidal E, Dasti A, Segura-Morales C, De Andrés-Aguayo L, Gómez A, Goldmann J, Jaenisch R, and Graf T
- Subjects
- Animals, Cell Differentiation genetics, Cell Lineage, Embryonic Stem Cells cytology, Germ Layers cytology, Mice, Neural Plate cytology, Nuclear Proteins metabolism, Transcription Factors metabolism, Cell Differentiation physiology, Endoderm cytology, Histone-Lysine N-Methyltransferase metabolism, Pluripotent Stem Cells cytology
- Abstract
How pluripotent stem cells differentiate into the main germ layers is a key question of developmental biology. Here, we show that the chromatin-related factor Whsc1 (also known as Nsd2 and MMSET) has a dual role in pluripotency exit and germ layer specification of embryonic stem cells. On induction of differentiation, a proportion of Whsc1-depleted embryonic stem cells remain entrapped in a pluripotent state and fail to form mesendoderm, although they are still capable of generating neuroectoderm. These functions of Whsc1 are independent of its methyltransferase activity. Whsc1 binds to enhancers of the mesendodermal regulators Gata4, T (Brachyury), Gata6 and Foxa2, together with Brd4, and activates the expression of these genes. Depleting each of these regulators also delays pluripotency exit, suggesting that they mediate the effects observed with Whsc1. Our data indicate that Whsc1 links silencing of the pluripotency regulatory network with activation of mesendoderm lineages.
- Published
- 2019
- Full Text
- View/download PDF
24. Transcription Factors Drive Tet2-Mediated Enhancer Demethylation to Reprogram Cell Fate.
- Author
-
Sardina JL, Collombet S, Tian TV, Gómez A, Di Stefano B, Berenguer C, Brumbaugh J, Stadhouders R, Segura-Morales C, Gut M, Gut IG, Heath S, Aranda S, Di Croce L, Hochedlinger K, Thieffry D, and Graf T
- Published
- 2018
- Full Text
- View/download PDF
25. TMPRSS2:ERG gene fusion expression regulates bone markers and enhances the osteoblastic phenotype of prostate cancer bone metastases.
- Author
-
Delliaux C, Tian TV, Bouchet M, Fradet A, Vanpouille N, Flourens A, Deplus R, Villers A, Leroy X, Clézardin P, de Launoit Y, Bonnelye E, and Duterque-Coquillaud M
- Subjects
- Alkaline Phosphatase genetics, Alkaline Phosphatase metabolism, Animals, Biomarkers, Tumor metabolism, Bone Neoplasms metabolism, Bone Neoplasms secondary, Cell Line, Tumor, Collagen Type I, alpha 1 Chain, Endothelin-1 genetics, Endothelin-1 metabolism, Humans, Male, Mice, SCID, Oncogene Proteins, Fusion metabolism, Osteoblasts pathology, PC-3 Cells, Phenotype, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Transplantation, Heterologous, Tumor Burden genetics, Biomarkers, Tumor genetics, Bone Neoplasms genetics, Gene Expression Regulation, Neoplastic, Oncogene Proteins, Fusion genetics, Osteoblasts metabolism, Prostatic Neoplasms genetics
- Abstract
Prostate cancers have a strong propensity to metastasize to bone and promote osteoblastic lesions. TMPRSS2:ERG is the most frequent gene rearrangement identified in prostate cancer, but whether it is involved in prostate cancer bone metastases is largely unknown. We exploited an intratibial metastasis model to address this issue and we found that ectopic expression of the TMPRSS2:ERG fusion enhances the ability of prostate cancer cell lines to induce osteoblastic lesions by stimulating bone formation and inhibiting the osteolytic response. In line with these in vivo results, we demonstrate that the TMPRSS2:ERG fusion protein increases the expression of osteoblastic markers, including Collagen Type I Alpha 1 Chain and Alkaline Phosphatase, as well as Endothelin-1, a protein with a documented role in osteoblastic bone lesion formation. Moreover, we determined that the TMPRSS2:ERG fusion protein is bound to the regulatory regions of these genes in prostate cancer cell lines, and we report that the expression levels of these osteoblastic markers are correlated with the expression of the TMPRSS2:ERG fusion in patient metastasis samples. Taken together, our results reveal that the TMPRSS2:ERG gene fusion is involved in osteoblastic lesion formation induced by prostate cancer cells., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
26. Constitutively Active SMAD2/3 Are Broad-Scope Potentiators of Transcription-Factor-Mediated Cellular Reprogramming.
- Author
-
Ruetz T, Pfisterer U, Di Stefano B, Ashmore J, Beniazza M, Tian TV, Kaemena DF, Tosti L, Tan W, Manning JR, Chantzoura E, Ottosson DR, Collombet S, Johnsson A, Cohen E, Yusa K, Linnarsson S, Graf T, Parmar M, and Kaji K
- Subjects
- Cell Line, Humans, Transcription Factors genetics, Cellular Reprogramming, Induced Pluripotent Stem Cells metabolism, Smad2 Protein metabolism, Smad3 Protein metabolism, Transcription Factors metabolism
- Abstract
Reprogramming of cellular identity using exogenous expression of transcription factors (TFs) is a powerful and exciting tool for tissue engineering, disease modeling, and regenerative medicine. However, generation of desired cell types using this approach is often plagued by inefficiency, slow conversion, and an inability to produce mature functional cells. Here, we show that expression of constitutively active SMAD2/3 significantly improves the efficiency of induced pluripotent stem cell (iPSC) generation by the Yamanaka factors. Mechanistically, SMAD3 interacts with reprogramming factors and co-activators and co-occupies OCT4 target loci during reprogramming. Unexpectedly, active SMAD2/3 also markedly enhances three other TF-mediated direct reprogramming conversions, from B cells to macrophages, myoblasts to adipocytes, and human fibroblasts to neurons, highlighting broad and general roles for SMAD2/3 as cell-reprogramming potentiators. Our results suggest that co-expression of active SMAD2/3 could enhance multiple types of TF-based cell identity conversion and therefore be a powerful tool for cellular engineering., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
27. ΔNp63α promotes adhesion of metastatic prostate cancer cells to the bone through regulation of CD82.
- Author
-
Di Giacomo V, Tian TV, Mas A, Pecoraro M, Batlle-Morera L, Noya L, Martín-Caballero J, Ruberte J, and Keyes WM
- Subjects
- Animals, Cell Adhesion, Cell Line, Tumor, Gene Expression Regulation, Humans, Male, Mice, Mice, Inbred C57BL, Bone Neoplasms secondary, Kangai-1 Protein physiology, Prostatic Neoplasms pathology, Transcription Factors physiology, Tumor Suppressor Proteins physiology
- Abstract
ΔNp63α is a critical mediator of epithelial development and stem cell function in a variety of tissues including the skin and breast, while overexpression of ΔNp63α acts as an oncogene to drive tumor formation and cancer stem cell properties in squamous cell carcinoma. However, with regards to the prostate, while ΔNp63α is expressed in the basal stem cells of the mature gland, during adenocarcinoma development, its expression is lost and its absence is used to clinically diagnose the malignant state. Surprisingly, here we identify a sub-population of bone metastatic prostate cancer cells in the PC3 cell line that express ΔNp63α. Interestingly, we discovered that ΔNp63α favors adhesion and stem-like growth of these cells in the bone microenvironment. In addition, we show that these properties require expression of the target gene CD82. Together, this work uncovers a population of bone metastatic prostate cancer cells that express ΔNp63α, and provides important information about the mechanisms of bone metastatic colonization. Finally, we identify metastasis-promoting properties for the tetraspanin family member CD82.
- Published
- 2017
- Full Text
- View/download PDF
28. Identification of novel TMPRSS2:ERG mechanisms in prostate cancer metastasis: involvement of MMP9 and PLXNA2.
- Author
-
Tian TV, Tomavo N, Huot L, Flourens A, Bonnelye E, Flajollet S, Hot D, Leroy X, de Launoit Y, and Duterque-Coquillaud M
- Subjects
- Cell Line, Tumor, Cell Movement, Cell Proliferation, Gene Expression Regulation, Humans, Lymphatic Metastasis, Male, Oncogene Proteins, Fusion genetics, Phenotype, Prostatic Neoplasms pathology, Transcriptome, Matrix Metalloproteinase 9 metabolism, Nerve Tissue Proteins metabolism, Oncogene Proteins, Fusion metabolism, Prostatic Neoplasms metabolism, Receptors, Cell Surface metabolism
- Abstract
Prostate cancer (PCa) is one of the major public health problems in Western countries. Recently, the TMPRSS2:ERG gene fusion, which results in the aberrant expression of the transcription factor ERG, has been shown to be the most common gene rearrangement in PCa. Previous studies have determined the contributions of this fusion in PCa disease initiation and/or progression in vitro and in vivo. In this study on TMPRSS2:ERG regulation in PCa, we used an androgen receptor and TMPRSS2:ERG fusion double-negative PCa cell model: PC3c. In three cell clones with different TMPRSS2:ERG expression levels, ectopic expression of the fusion resulted in significant induction of cell migration and invasion in a dose-dependent manner. In agreement with this phenotype, high-throughput microarray analysis revealed that a set of genes, functionally associated with cell motility and invasiveness, were deregulated in a dose-dependent manner in TMPRSS2:ERG-expressing cells. Importantly, we identified increased MMP9 (Metalloproteinase 9) and PLXNA2 (Plexin A2) expression in TMPRSS2:ERG-positive PCa samples, and their expression levels were significantly correlated with ERG expression in a PCa cohort. In line with these findings, there was evidence that TMPRSS2:ERG directly and positively regulates MMP9 and PLXNA2 expression in PC3c cells. Moreover, PLXNA2 upregulation contributed to TMPRSS2:ERG-mediated enhancements of PC3c cell migration and invasion. Furthermore, and importantly, PLXNA2 expression was upregulated in metastatic PCa tumors compared with localized primary PCa tumors. This study provides novel insights into the role of the TMPRSS2:ERG fusion in PCa metastasis.
- Published
- 2014
- Full Text
- View/download PDF
29. Increased adipogenesis in cultured embryonic chondrocytes and in adult bone marrow of dominant negative Erg transgenic mice.
- Author
-
Flajollet S, Tian TV, Huot L, Tomavo N, Flourens A, Holder-Espinasse M, Le Jeune M, Dumont P, Hot D, Mallein-Gerin F, and Duterque-Coquillaud M
- Subjects
- Animals, Bone Marrow Cells cytology, Cartilage cytology, Cartilage metabolism, Chondrocytes cytology, Mice, Mice, Transgenic, Proto-Oncogene Proteins c-ets metabolism, Adipogenesis genetics, Bone Marrow Cells metabolism, Chondrocytes metabolism, Chondrogenesis genetics, Proto-Oncogene Proteins c-ets genetics
- Abstract
In monolayer culture, primary articular chondrocytes have an intrinsic tendency to lose their phenotype during expansion. The molecular events underlying this chondrocyte dedifferentiation are still largely unknown. Several transcription factors are important for chondrocyte differentiation. The Ets transcription factor family may be involved in skeletal development. One family member, the Erg gene, is mainly expressed during cartilage formation. To further investigate the potential role of Erg in the maintenance of the chondrocyte phenotype, we isolated and cultured chondrocytes from the rib cartilage of embryos of transgenic mice that express a dominant negative form of Erg (DN-Erg) during cartilage formation. DN-Erg expression in chondrocytes cultured for up to 20 days did not affect the early dedifferentiation usually observed in cultured chondrocytes. However, lipid droplets accumulated in DN-Erg chondrocytes, suggesting adipocyte emergence. Transcriptomic analysis using a DNA microarray, validated by quantitative RT-PCR, revealed strong differential gene expression, with a decrease in chondrogenesis-related markers and an increase in adipogenesis-related gene expression in cultured DN-Erg chondrocytes. These results indicate that Erg is involved in either maintaining the chondrogenic phenotype in vitro or in cell fate orientation. Along with the in vitro studies, we compared adipocyte presence in wild-type and transgenic mice skeletons. Histological investigations revealed an increase in the number of adipocytes in the bone marrow of adult DN-Erg mice even though no adipocytes were detected in embryonic cartilage or bone. These findings suggest that the Ets transcription factor family may contribute to the homeostatic balance in skeleton cell plasticity.
- Published
- 2012
- Full Text
- View/download PDF
30. Abnormal expression of the ERG transcription factor in prostate cancer cells activates osteopontin.
- Author
-
Flajollet S, Tian TV, Flourens A, Tomavo N, Villers A, Bonnelye E, Aubert S, Leroy X, and Duterque-Coquillaud M
- Subjects
- Aged, Binding Sites, Cell Line, Tumor, Female, HeLa Cells, Humans, Male, Middle Aged, Neoplasm Metastasis, Promoter Regions, Genetic, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Transcriptional Regulator ERG, Gene Expression Regulation, Neoplastic, Oncogene Proteins, Fusion metabolism, Osteopontin genetics, Prostatic Neoplasms pathology, Trans-Activators metabolism, Transcriptional Activation
- Abstract
Osteopontin (OPN) is an extracellular matrix glycophosphoprotein that plays a key role in the metastasis of a wide variety of cancers. The high level of OPN expression in prostate cells is associated with malignancy and reduced survival of the patient. Recent studies on prostate cancer (PCa) tissue have revealed recurrent genomic rearrangements involving the fusion of the 5' untranslated region of a prostate-specific androgen-responsive gene with a gene coding for transcription factors from the ETS family. The most frequently identified fusion gene is TMPRSS2:ERG, which causes ERG protein overexpression in PCa cells. ERG is a transcription factor linked to skeletogenesis. This study was designed to test whether ERG and the product of the TMPRSS2:ERG fusion gene modulate OPN gene expression in PCa cells. To characterize ERG and TMPRSS2:ERG transcriptional activity of OPN, we focused on ETS binding sites (EBS) localized in conserved regions of the promoter. Using in vitro and in vivo molecular assays, we showed that ERG increases OPN expression and binds to an EBS (nt -115 to -118) in the OPN promoter. Moreover, stable transfection of prostate tumor cell lines by TMPRSS2:ERG upregulates endogenous OPN expression. Finally, in human prostate tumor samples, detection of the TMPRSS2:ERG fusion gene was significantly associated with OPN overexpression. Taken together, these data suggest that OPN is an ERG-target gene in PCa where the abnormal expression of the transcription factor ERG, due to the TMPRSS2:ERG fusion, disturbs the expression of genes that play an important role in PCa cells and associated metastases., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
31. Identification of four alternatively spliced transcripts of the Ucma/GRP gene, encoding a new Gla-containing protein.
- Author
-
Le Jeune M, Tomavo N, Tian TV, Flourens A, Marchand N, Camuzeaux B, Mallein-Gerin F, and Duterque-Coquillaud M
- Subjects
- Animals, Bone Morphogenetic Protein 2 pharmacology, Cell Differentiation physiology, Chondrocytes metabolism, Chondrogenesis physiology, Cytoplasm metabolism, Embryo, Mammalian cytology, Embryo, Mammalian metabolism, Exons genetics, Extracellular Matrix Proteins, Gene Expression drug effects, Gene Expression genetics, Gene Expression Regulation, Developmental physiology, Golgi Apparatus metabolism, HeLa Cells, Humans, Intracellular Signaling Peptides and Proteins, Mice, Mice, Transgenic, Microtubules drug effects, Microtubules metabolism, Molecular Sequence Data, Nocodazole pharmacology, Organelles metabolism, Protein Isoforms chemistry, Protein Isoforms genetics, Protein Isoforms metabolism, Proteins chemistry, Proto-Oncogene Protein c-fli-1 genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Transforming Growth Factor beta1 pharmacology, 1-Carboxyglutamic Acid analysis, Alternative Splicing genetics, Proteins genetics
- Abstract
The Ucma protein (Upper zone of growth plate and cartilage matrix associated protein) has recently been described as a novel secretory protein mainly expressed in cartilage and also as a novel vitamin-K-dependent protein named GRP (Gla-rich protein). This protein has the highest Gla content of any protein known to date. In this article, we identify four alternatively spliced variants of Ucma/GRP gene transcripts in mouse chondrocytes. We show that the expression of all four isoforms is associated with the early stages of chondrogenesis. The Ucma/GRP gene encodes four proteins named Ucma/GRP-F1, -F2, -F3, and -F4, which differ by exon 2, exon 4, or both. Among them, only Ucma/GRP-F1 and -F3 were secreted into the culture medium of transfected chondrocytes, while Ucma/GRP-F2 and -F4 accumulated in the cells. Using HeLa cells or freshly isolated embryonic mouse chondrocytes transfected with enhanced green fluorescent protein fusions, microscopy analysis revealed that Ucma/GRP-F1 and -F3 were localized in the Golgi complex, whereas Ucma/GRP-F2 and -F4 formed aggregates. This aggregation was microtubule-dependent since disruption of microtubules with nocodazole reduced Ucma/GRP-F2 and -F4 aggregation in a reversible manner. Using biochemical fractionation and Western blot analysis, Ucma/GRP-F1 and -F3 isoforms were detected in the soluble fraction while Ucma/GRP-F2 and -F4 were found in an insoluble-enriched fraction. We conclude that the co-expression of soluble and insoluble isoforms also Gla-rich and Gla-deleted isoforms may be finely tuned. Imbalance in isoform expression may therefore be involved in skeletal pathology.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.