158 results on '"Ludovic Vallier"'
Search Results
2. FXR inhibition may protect from SARS-CoV-2 infection by reducing ACE2
- Author
-
Teresa, Brevini, Mailis, Maes, Gwilym J, Webb, Binu V, John, Claudia D, Fuchs, Gustav, Buescher, Lu, Wang, Chelsea, Griffiths, Marnie L, Brown, William E, Scott, Pehuén, Pereyra-Gerber, William T H, Gelson, Stephanie, Brown, Scott, Dillon, Daniele, Muraro, Jo, Sharp, Megan, Neary, Helen, Box, Lee, Tatham, James, Stewart, Paul, Curley, Henry, Pertinez, Sally, Forrest, Petra, Mlcochova, Sagar S, Varankar, Mahnaz, Darvish-Damavandi, Victoria L, Mulcahy, Rhoda E, Kuc, Thomas L, Williams, James A, Heslop, Davide, Rossetti, Olivia C, Tysoe, Vasileios, Galanakis, Marta, Vila-Gonzalez, Thomas W M, Crozier, Johannes, Bargehr, Sanjay, Sinha, Sara S, Upponi, Corrina, Fear, Lisa, Swift, Kourosh, Saeb-Parsy, Susan E, Davies, Axel, Wester, Hannes, Hagström, Espen, Melum, Darran, Clements, Peter, Humphreys, Jo, Herriott, Edyta, Kijak, Helen, Cox, Chloe, Bramwell, Anthony, Valentijn, Christopher J R, Illingworth, Bassam, Dahman, Dustin R, Bastaich, Raphaella D, Ferreira, Thomas, Marjot, Eleanor, Barnes, Andrew M, Moon, Alfred S, Barritt, Ravindra K, Gupta, Stephen, Baker, Anthony P, Davenport, Gareth, Corbett, Vassilis G, Gorgoulis, Simon J A, Buczacki, Joo-Hyeon, Lee, Nicholas J, Matheson, Michael, Trauner, Andrew J, Fisher, Paul, Gibbs, Andrew J, Butler, Christopher J E, Watson, George F, Mells, Gordon, Dougan, Andrew, Owen, Ansgar W, Lohse, Ludovic, Vallier, Fotios, Sampaziotis, and consortium, UK-PBC research
- Subjects
Multidisciplinary - Abstract
Preventing SARS-CoV-2 infection by modulating viral host receptors, such as angiotensin-converting enzyme 2 (ACE2)1, could represent a new chemoprophylactic approach for COVID-19 that complements vaccination2,3. However, the mechanisms that control the expression of ACE2 remain unclear. Here we show that the farnesoid X receptor (FXR) is a direct regulator of ACE2 transcription in several tissues affected by COVID-19, including the gastrointestinal and respiratory systems. We then use the over-the-counter compound z-guggulsterone and the off-patent drug ursodeoxycholic acid (UDCA) to reduce FXR signalling and downregulate ACE2 in human lung, cholangiocyte and intestinal organoids and in the corresponding tissues in mice and hamsters. We show that the UDCA-mediated downregulation of ACE2 reduces susceptibility to SARS-CoV-2 infection in vitro, in vivo and in human lungs and livers perfused ex situ. Furthermore, we reveal that UDCA reduces the expression of ACE2 in the nasal epithelium in humans. Finally, we identify a correlation between UDCA treatment and positive clinical outcomes after SARS-CoV-2 infection using retrospective registry data, and confirm these findings in an independent validation cohort of recipients of liver transplants. In conclusion, we show that FXR has a role in controlling ACE2 expression and provide evidence that modulation of this pathway could be beneficial for reducing SARS-CoV-2 infection, paving the way for future clinical trials.
- Published
- 2022
3. Single-cell atlas of human liver development reveals pathways directing hepatic cell fates
- Author
-
Brandon T. Wesley, Alexander D. B. Ross, Daniele Muraro, Zhichao Miao, Sarah Saxton, Rute A. Tomaz, Carola M. Morell, Katherine Ridley, Ekaterini D. Zacharis, Sandra Petrus-Reurer, Judith Kraiczy, Krishnaa T. Mahbubani, Stephanie Brown, Jose Garcia-Bernardo, Clara Alsinet, Daniel Gaffney, Dave Horsfall, Olivia C. Tysoe, Rachel A. Botting, Emily Stephenson, Dorin-Mirel Popescu, Sonya MacParland, Gary Bader, Ian D. McGilvray, Daniel Ortmann, Fotios Sampaziotis, Kourosh Saeb-Parsy, Muzlifah Haniffa, Kelly R. Stevens, Matthias Zilbauer, Sarah A. Teichmann, and Ludovic Vallier
- Subjects
Organoids ,Liver ,Hepatocytes ,Humans ,Cell Differentiation ,Cell Biology ,Transcription Factors - Abstract
The liver has been studied extensively due to the broad number of diseases affecting its vital functions. However, therapeutic advances have been hampered by the lack of knowledge concerning human hepatic development. Here, we addressed this limitation by describing the developmental trajectories of different cell types that make up the human liver at single-cell resolution. These transcriptomic analyses revealed that sequential cell-to-cell interactions direct functional maturation of hepatocytes, with non-parenchymal cells playing essential roles during organogenesis. We utilized this information to derive bipotential hepatoblast organoids and then exploited this model system to validate the importance of signalling pathways in hepatocyte and cholangiocyte specification. Further insights into hepatic maturation also enabled the identification of stage-specific transcription factors to improve the functionality of hepatocyte-like cells generated from human pluripotent stem cells. Thus, our study establishes a platform to investigate the basic mechanisms directing human liver development and to produce cell types for clinical applications.
- Published
- 2022
4. Assessing motile cilia coverage and beat frequency in mammalian in-vitro cell culture tissues
- Author
-
Ricardo Fradique, Erika Causa, Clara Delahousse, Jurij Kotar, Laetitia Pinte, Ludovic Vallier, Marta Vila-Gonzalez, and Pietro Cicuta
- Abstract
Cilia density, distribution and beating frequency are important parameters lung tissues, for example in diagnostics of Primary Ciliary Dyskinesia, and in the study ofin vitromodels, e.g. derived from induced Pluripotent Stem Cells. Video microscopy can be used to characterise these parameters, but most tools available at the moment are limited in the type of information they can provide, usually only describing the ciliary beat frequency of very small areas, while requiring human intervention and training for their use. We propose a novel and open source method to fully characterise cilia beating frequency and motile cilia coverage in an automated fashion without user intervention. We demonstrate the ability to differentiate between different coverage densities, identifying even small patches of cilia in a larger field of view, and to fully characterise the cilia beating frequency of all moving areas. We also show that the method can be used to combine multiple fields of view to better describe a sample without relying on small pre-selected regions of interest. This is released with a simple graphical user interface for file handling, enabling a full analysis of individual fields of view in a few minutes on a typical personal computer.
- Published
- 2023
5. Human branching cholangiocyte organoids recapitulate functional bile duct formation
- Author
-
Floris J.M. Roos, Gilles S. van Tienderen, Haoyu Wu, Ignacio Bordeu, Dina Vinke, Laura Muñoz Albarinos, Kathryn Monfils, Sabrah Niesten, Ron Smits, Jorke Willemse, Oskar Rosmark, Gunilla Westergren-Thorsson, Daniel J. Kunz, Maurice de Wit, Pim J. French, Ludovic Vallier, Jan N.M. IJzermans, Richard Bartfai, Hendrik Marks, Ben D. Simons, Martin E. van Royen, Monique M.A. Verstegen, Luc J.W. van der Laan, Surgery, Gastroenterology & Hepatology, Pathology, Neurology, Roos, Floris Johan Maria [0000-0003-1278-6517], Kunz, Daniel [0000-0003-3597-6591], Vallier, Ludovic [0000-0002-3848-2602], Simons, Benjamin [0000-0002-3875-7071], and Apollo - University of Cambridge Repository
- Subjects
Epithelial Cells ,Cell Biology ,Cholangiocarcinoma ,Organoids ,cholangiocyte organoids ,SDG 3 - Good Health and Well-being ,embryonic bile duct development ,disease modeling ,Genetics ,Molecular Medicine ,Humans ,Bile Ducts ,branching morphogenesis ,cholangiocytes ,Transcriptome ,Molecular Biology ,intrahepatic bile duct - Abstract
Human cholangiocyte organoids show great promise for regenerative therapies and in vitro modeling of bile duct development and diseases. However, the cystic organoids lack the branching morphology of intrahepatic bile ducts (IHBDs). Here, we report establishing human branching cholangiocyte organoid (BRCO) cultures. BRCOs self-organize into complex tubular structures resembling the IHBD architecture. Single-cell transcriptomics and functional analysis showed high similarity to primary cholangiocytes, and importantly, the branching growth mimics aspects of tubular development and is dependent on JAG1/NOTCH2 signaling. When applied to cholangiocarcinoma tumor organoids, the morphology changes to an in vitro morphology like primary tumors. Moreover, these branching cholangiocarcinoma organoids (BRCCAOs) better match the transcriptomic profile of primary tumors and showed increased chemoresistance to gemcitabine and cisplatin. In conclusion, BRCOs recapitulate a complex process of branching morphogenesis in vitro. This provides an improved model to study tubular formation, bile duct functionality, and associated biliary diseases.
- Published
- 2023
- Full Text
- View/download PDF
6. A single-cell comparison of adult and fetal human epicardium defines the age-associated changes in epicardial activity
- Author
-
Vincent R. Knight-Schrijver, Hongorzul Davaapil, Semih Bayraktar, Alexander D. B. Ross, Kazumasa Kanemaru, James Cranley, Monika Dabrowska, Minal Patel, Krzysztof Polanski, Xiaoling He, Ludovic Vallier, Sarah Teichmann, Laure Gambardella, Sanjay Sinha, Knight-Schrijver, Vincent R [0000-0002-7916-3827], Gambardella, Laure [0000-0001-5771-1565], Sinha, Sanjay [0000-0001-5900-1209], Apollo - University of Cambridge Repository, and Knight-Schrijver, Vincent R. [0000-0002-7916-3827]
- Subjects
Resource ,14/1 ,631/532/7 ,631/136/532/489 ,631/208/514/1949 ,45 ,13/100 ,Cambridge Stem Cell Institute ,82 ,692/308/2171 ,631/443/592/2726 ,38/91 ,38 - Abstract
Acknowledgements: The authors would like to thank R. Barker at the University of Cambridge for assistance in obtaining the fetal tissue samples used in all analyses. Additionally, we are grateful for support from the Wellcome Sanger Cellular Generation and Phenotyping team and the Core DNA Pipelines team. This research was funded by the British Heart Foundation (BHF) Senior Fellowship (FS/18/46/33663)(S.S. and L.G.); the Oxbridge BHF Centre for Regenerative Medicine (RM/17/2/33380) (V.K.S.); and BHF grants PG/17/24/32886 (L.G.) and RG/17/5/32936 (H.D.). We also acknowledge core support from the Wellcome Trust, the Medical Research Council and the Wellcome Trust–Medical Research Council Cambridge Stem Cell Institute. This research was funded, in whole or in part, by the Wellcome Trust (grant no. 203151/Z/16/Z). Finally, this project has been made possible, in part, by the Wellcome Trust (WT206194, S.A.T), the Wellcome Trust Clinical PhD Fellowship (J.C) and the Overseas Research Fellowship of the Takeda Science Foundation (K.K). For the purpose of open access, the author has applied a CC BY public copyright licence to any Author Accepted Manuscript version arising from this submission., Funder: Takeda Science Foundation; doi: https://doi.org/10.13039/100007449, Funder: Wellcome Trust Clinical PhD Fellowship, Re-activating quiescent adult epicardium represents a potential therapeutic approach for human cardiac regeneration. However, the exact molecular differences between inactive adult and active fetal epicardium are not known. In this study, we combined fetal and adult human hearts using single-cell and single-nuclei RNA sequencing and compared epicardial cells from both stages. We found that a migratory fibroblast-like epicardial population only in the fetal heart and fetal epicardium expressed angiogenic gene programs, whereas the adult epicardium was solely mesothelial and immune responsive. Furthermore, we predicted that adult hearts may still receive fetal epicardial paracrine communication, including WNT signaling with endocardium, reinforcing the validity of regenerative strategies that administer or reactivate epicardial cells in situ. Finally, we explained graft efficacy of our human embryonic stem-cell-derived epicardium model by noting its similarity to human fetal epicardium. Overall, our study defines epicardial programs of regenerative angiogenesis absent in adult hearts, contextualizes animal studies and defines epicardial states required for effective human heart regeneration.
- Published
- 2022
7. Single-cell transcriptomic characterization of a gastrulating human embryo
- Author
-
Elmir Mahammadov, Shota Nakanoh, Shankar Srinivas, Ludovic Vallier, Antonio Scialdone, Richard C. V. Tyser, Tyser, Richard CV [0000-0001-7884-1756], Mahammadov, Elmir [0000-0003-0008-1413], Vallier, Ludovic [0000-0002-3848-2602], Scialdone, Antonio [0000-0002-4956-2843], Srinivas, Shankar [0000-0001-5726-7791], and Apollo - University of Cambridge Repository
- Subjects
Male ,Mesoderm ,Cell type ,Erythrocytes ,Datasets as Topic ,Context (language use) ,Biology ,Mice ,Directed differentiation ,medicine ,Animals ,Humans ,Induced pluripotent stem cell ,Multidisciplinary ,Gene Expression Profiling ,Endoderm ,Gastrulation ,Cell Differentiation ,Embryo ,Gastrula ,Embryo, Mammalian ,Cell biology ,Germ Cells ,medicine.anatomical_structure ,Epiblast ,Female ,Single-Cell Analysis ,Transcriptome - Abstract
Gastrulation is the fundamental process in all multicellular animals through which the basic body plan is first laid down1–4. It is pivotal in generating cellular diversity coordinated with spatial patterning. In humans, gastrulation occurs in the third week after fertilization. Our understanding of this process in humans is relatively limited and based primarily on historical specimens5–8, experimental models9–12 or, more recently, in vitro cultured samples13–16. Here we characterize in a spatially resolved manner the single-cell transcriptional profile of an entire gastrulating human embryo, staged to be between 16 and 19 days after fertilization. We use these data to analyse the cell types present and to make comparisons with other model systems. In addition to pluripotent epiblast, we identified primordial germ cells, red blood cells and various mesodermal and endodermal cell types. This dataset offers a unique glimpse into a central but inaccessible stage of our development. This characterization provides new context for interpreting experiments in other model systems and represents a valuable resource for guiding directed differentiation of human cells in vitro. The single-cell transcriptional profile of a human embryo between 16 and 19 days after fertilization reveals parallels and differences in gastrulation in humans as compared with mouse and non-human primate models.
- Published
- 2021
8. Limited oxygen availability in standard cell culture alters metabolism and function in terminally-differentiated cells
- Author
-
Joycelyn Tan, Sam Virtue, Dougall M. Norris, Olivia J. Conway, Ming Yang, Christopher Gribben, Fatima Lugtu, Ioannis Kamzolas, James R. Krycer, Richard J. Mills, Conceição Pereira, Martin Dale, Amber S. Shun-Shion, Harry J. M. Baird, James A. Horscroft, Alice P. Sowton, Marcella Ma, Stefania Carobbio, Evangelia Petsalaki, Andrew J. Murray, David C. Gershlick, James E. Hudson, Ludovic Vallier, Kelsey H Fisher-Wellman, Christian Frezza, Antonio Vidal-Puig, and Daniel J. Fazakerley
- Abstract
SUMMARYCell culture is generally considered to be hyperoxic. However, the importance of cellular oxygen consumption is often underappreciated, with rates of oxygen consumption often sufficient to cause hypoxia at cell monolayers. We initially focused on cultured adipocytes as a terminally differentiated cell-type with substantial oxygen consumption rates to support diverse cellular functions. Under standard conditions, cultured adipocytes are hypoxic and highly glycolytic. Increasing oxygen diverted glucose flux toward mitochondria and resulted in thousands of gene expression changes that pointed toward alleviated physiological transcriptional responses to hypoxia. Phenotypically, providing more oxygen increased adipokine secretion and rendered adipocytes more sensitive to insulin and lipolytic stimuli. The functional benefits of increasing pericellular oxygen were transferable to other cellular systems including hPSC-derived hepatocytes and cardiac organoids. Our findings suggest that oxygen is limiting in many terminally-differentiated cell culture systems, and that controlling oxygen availability can improve the quality and translatability of cell models.
- Published
- 2022
9. Generation of functional hepatocytes by forward programming with nuclear receptors
- Author
-
Rute A. Tomaz, Ekaterini D. Zacharis, Fabian Bachinger, Annabelle Wurmser, Daniel Yamamoto, Sandra Petrus-Reurer, Carola M. Morell, Dominika Dziedzicka, Brandon T. Wesley, Imbisaat Geti, Charis-Patricia Segeritz, Miguel Cardoso de Brito, Mariya Chhatriwala, Daniel Ortmann, Kourosh Saeb-Parsy, Ludovic Vallier, Tomaz, Rute A [0000-0002-9377-1431], Vallier, Ludovic [0000-0002-3848-2602], and Apollo - University of Cambridge Repository
- Subjects
General Immunology and Microbiology ,General Neuroscience ,Induced Pluripotent Stem Cells ,regenerative medicine ,Receptors, Cytoplasmic and Nuclear ,Cell Differentiation ,General Medicine ,Stem Cells and Regenerative Medicine ,General Biochemistry, Genetics and Molecular Biology ,Tools and Resources ,Liver ,stem cells ,Humans ,hepatocytes ,pluripotent stem cells ,forward programming ,Human - Abstract
Funder: UK Regenerative Medicine Platform, Funder: Wellcome Trust, Funder: Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Funder: Chan Zuckerberg Initiative, Production of large quantities of hepatocytes remains a major challenge for a number of clinical applications in the biomedical field. Directed differentiation of human pluripotent stem cells (hPSCs) into hepatocyte-like cells (HLCs) provides an advantageous solution and a number of protocols have been developed for this purpose. However, these methods usually follow different steps of liver development in vitro, which is time consuming and requires complex culture conditions. In addition, HLCs lack the full repertoire of functionalities characterising primary hepatocytes. Here, we explore the interest of forward programming to generate hepatocytes from hPSCs and to bypass these limitations. This approach relies on the overexpression of three hepatocyte nuclear factors (HNF1A, HNF6, and FOXA3) in combination with different nuclear receptors expressed in the adult liver using the OPTi-OX platform. Forward programming allows for the rapid production of hepatocytes (FoP-Heps) with functional characteristics using a simplified process. We also uncovered that the overexpression of nuclear receptors such as RORc can enhance specific functionalities of FoP-Heps thereby validating its role in lipid/glucose metabolism. Together, our results show that forward programming could offer a versatile alternative to direct differentiation for generating hepatocytes in vitro.
- Published
- 2022
- Full Text
- View/download PDF
10. Author response: Generation of functional hepatocytes by forward programming with nuclear receptors
- Author
-
Rute A Tomaz, Ekaterini D Zacharis, Fabian Bachinger, Annabelle Wurmser, Daniel Yamamoto, Sandra Petrus-Reurer, Carola M Morell, Dominika Dziedzicka, Brandon T Wesley, Imbisaat Geti, Charis-Patricia Segeritz, Miguel C de Brito, Mariya Chhatriwala, Daniel Ortmann, Kourosh Saeb-Parsy, and Ludovic Vallier
- Published
- 2022
11. Author Correction: A single-cell comparison of adult and fetal human epicardium defines the age-associated changes in epicardial activity
- Author
-
Vincent R. Knight-Schrijver, Hongorzul Davaapil, Semih Bayraktar, Alexander D. B. Ross, Kazumasa Kanemaru, James Cranley, Monika Dabrowska, Minal Patel, Krzysztof Polanski, Xiaoling He, Ludovic Vallier, Sarah Teichmann, Laure Gambardella, and Sanjay Sinha
- Published
- 2023
12. Single-cell atlas of human liver development reveals pathways directing hepatic cell fates
- Author
-
Brandon T. Wesley, Alexander D. B. Ross, Daniele Muraro, Zhichao Miao, Sarah Saxton, Rute A. Tomaz, Carola M. Morell, Katherine Ridley, Ekaterini D. Zacharis, Sandra Petrus-Reurer, Judith Kraiczy, Krishnaa T. Mahbubani, Stephanie Brown, Jose Garcia-Bernardo, Clara Alsinet, Daniel Gaffney, Olivia C. Tysoe, Rachel A. Botting, Emily Stephenson, Dorin-Mirel Popescu, Sonya MacParland, Gary Bader, Ian D. McGilvray, Daniel Ortmann, Fotios Sampaziotis, Kourosh Saeb-Parsy, Muzlifah Haniffa, Kelly R. Stevens, Matthias Zilbauer, Sarah A. Teichmann, and Ludovic Vallier
- Abstract
The liver has been studied extensively due to the broad number of diseases affecting its vital functions. However, therapeutic advances, especially in regenerative medicine, are currently hampered by the lack of knowledge concerning human hepatic cell development. Here, we addressed this limitation by describing the developmental trajectories of different cell types comprising the human fetal liver at single-cell resolution. These transcriptomic analyses revealed that sequential cell-to-cell interactions direct functional maturation of hepatocytes, with non-parenchymal cells playing critical, supportive roles during organogenesis. We utilised this information to derive bipotential hepatoblast organoids and then exploited this novel model system to validate the importance of key signalling pathways and developmental cues. Furthermore, these insights into hepatic maturation enabled the identification of stage-specific transcription factors to improve the functionality of hepatocyte-like cells generated from human pluripotent stem cells. Thus, our study establishes a new platform to investigate the basic mechanisms of human liver development and to produce cell types for clinical applications.
- Published
- 2022
13. A p53-Dependent Checkpoint Induced upon DNA Damage Alters Cell Fate during hiPSC Differentiation
- Author
-
Julian E. Sale, Alastair Crisp, Cara B. Eldridge, Finian J. Allen, Rodrigo A. Grandy, Ludovic Vallier, Allen, Finian [0000-0002-9823-562X], Vallier, Ludovic [0000-0002-3848-2602], and Apollo - University of Cambridge Repository
- Subjects
p53 ,0301 basic medicine ,Mesoderm ,Transcription, Genetic ,DNA damage ,Induced Pluripotent Stem Cells ,Germ layer ,Biology ,Cell fate determination ,Biochemistry ,hiPSC ,03 medical and health sciences ,checkpoint ,0302 clinical medicine ,Tissue engineering ,Report ,Genetics ,medicine ,Humans ,Cell Lineage ,Endoderm ,DNA-damage response ,Cell Differentiation ,differentiation ,Cell Cycle Checkpoints ,Cell Biology ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,hESC ,Apoptosis ,definitive endoderm ,Tumor Suppressor Protein p53 ,030217 neurology & neurosurgery ,DNA Damage ,Developmental Biology ,Definitive endoderm - Abstract
Summary The ability of human induced pluripotent stem cells (hiPSCs) to differentiate in vitro to each of the three germ layer lineages has made them an important model of early human development and a tool for tissue engineering. However, the factors that disturb the intricate transcriptional choreography of differentiation remain incompletely understood. Here, we uncover a critical time window during which DNA damage significantly reduces the efficiency and fidelity with which hiPSCs differentiate to definitive endoderm. DNA damage prevents the normal reduction of p53 levels as cells pass through the epithelial-to-mesenchymal transition, diverting the transcriptional program toward mesoderm without induction of an apoptotic response. In contrast, TP53-deficient cells differentiate to endoderm with high efficiency after DNA damage, suggesting that p53 enforces a “differentiation checkpoint” in early endoderm differentiation that alters cell fate in response to DNA damage., Highlights • DNA damage impairs the efficiency and fidelity of human stem cell differentiation • p53 is reduced transiently during the commitment to definitive endoderm • Preventing p53 reduction diverts cells away from endoderm without apoptosis • p53-deficient cells differentiate to endoderm efficiently in the face of DNA damage, Eldridge et al. demonstrate a temporal window during which the differentiation of human iPSCs to definitive endoderm is sensitive to low levels of DNA damage, causing diversion of the differentiation program toward a more mesodermal fate. This effect is almost entirely dependent on p53, with both the efficiency and the fidelity of endoderm differentiation being restored in damaged p53-deficient cells.
- Published
- 2020
14. Regenerative and non-regenerative transcriptional states of the human epicardium: from foetus to adult and back again
- Author
-
Xiaoling He, Laure Gambardella, Sanjay Sinha, Ludovic Vallier, Vincent Knight-Schrijver, Hongorzul Davaapil, and Alexander Ross
- Subjects
Fetus ,education.field_of_study ,Cell type ,Angiogenesis ,Regeneration (biology) ,Population ,Context (language use) ,Biology ,Embryonic stem cell ,Cell biology ,embryonic structures ,cardiovascular system ,Animal studies ,education - Abstract
Epicardial activation appears to be required for cardiac regeneration. Although reverting quiescent adult epicardium to an active neonatal or foetal state will likely represent a key therapeutic approach for human cardiac regeneration, the exact molecular differences between human adult and foetal epicardium are not understood. We used single-cell RNA sequencing to compare epicardial cells from both foetal and adult hearts. We found two foetal epicardial cell types, mesothelial and fibroblast-like, with only the mesothelial population present in adults. We also identified foetal-specific epicardial genes associated with regeneration and angiogenesis, and found that adult epicardium may be primed for immune and inflammatory responses. We predict that restoring the foetal epicardial state in human hearts would increase adult angiogenic potential. Finally, we demonstrated that human embryonic stem-cell derived epicardium is a valid model for the foetal epicardium and for investigating epicardial-mediated cardiac regeneration in humans. Our study defines regenerative programs in human foetal epicardium that are absent in the adult, brings human context to animal studies, and provides a roadmap for directing the epicardium in human heart regeneration.
- Published
- 2021
15. O07 FXR antagonists as new agents for COVID19
- Author
-
Olivia C. Tysoe, A Butler, Daniele Muraro, T. Brevini, Alfred Barritt, Sara Upponi, Gordon Dougan, Eleanor Barnes, R. E. Kuc, V. L. Mulcahy, Sally Forrest, Andrew Davenport, S. J. A. Buczacki, T. L. Williams, Ludovic Vallier, Mailis Maes, Paul Gibbs, S. Varankar, R. K. Gupta, Ansgar W. Lohse, George F. Mells, S. Baker, Christopher J.E. Watson, Andrew M. Moon, Kourosh Saeb-Parsy, P. Mlcochova, Sanjay Sinha, L. Swift, T. W. M. Crozier, M. Darvish-Damavandi, Susan E. Davies, Johannes Bargehr, M. Vila-Gonzalez, Gwilym J. Webb, William Gelson, F. Sampaziotis, Thomas Marjot, V. Galanakis, J. H. Lee, S. Dillon, and Gareth Corbett
- Subjects
medicine.diagnostic_test ,business.industry ,Pharmacology ,Cholangiocyte ,Ursodeoxycholic acid ,Transplantation ,Clinical trial ,Bronchoalveolar lavage ,Downregulation and upregulation ,In vivo ,medicine ,Organoid ,business ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
Introduction The management of COVID19 is complicated by vaccine availability, the modest efficacy of existing treatments, and the potential for viral resistance. Therefore, there is a pressing need for new prophylactic and therapeutic agents. The viral receptor ACE2 is an ideal target as it is required for SARS-CoV-2 entry in host cells. Modifying ACE2 expression could prevent infection and/or limit disease progression. Nevertheless, the mechanisms controlling ACE2 expression remain elusive. Aims To identify pathways controlling the transcriptional regulation of ACE2, and exploit them to reduce SARS-CoV-2 infection. Methods Organoids from primary biliary, intestinal and pulmonary epithelia were derived and cultured as previously described. Single-cell RNA sequencing, QPCR, immunofluorescence and flow cytometry were used to assess marker expression. Chromatin immunoprecipitation was used to assess FXR binding on DNA. Bronchoalveolar lavage SARS-CoV-2 patient isolates were used for infection experiments. Human livers not used for transplantation were connected to the metra (OrganOx) normothermic perfusion device and perfused ex-situ using therapeutic doses of UDCA for 12 hours. ACE2 activity was measured following manufacturer's instructions. Patient data from the COVID-Hep and SECURE-Liver registries were compared using propensity score matching for sex, age and Child-Turcotte-Pugh score. Results We first demonstrated that cholangiocytes are susceptible to SARS-CoV-2 infection in vivo and in organoid culture. We then used cholangiocyte organoids to identify FXR as a transcriptional regulator of ACE2. We validated our results in pulmonary and intestinal organoids, showing that ACE2 regulation by FXR represents a broad mechanism present in multiple COVID19-affected tissues. We then demonstrated that approved FXR inhibitors, such as ursodeoxycholic acid (UDCA) and z-guggulsterone (ZGG), decrease ACE2 levels and reduce viral infection in vitro in primary biliary, intestinal and pulmonary organoids. We interrogated the impact of systemic UDCA administration in human livers perfused ex-situ, demonstrating reduced ACE2 levels and SARS-CoV-2 infection. Furthermore, we showed that commencing UDCA treatment lowers ACE2 levels in primary biliary cholangitis (PBC) patients. Finally, we identified a correlation between UDCA treatment and better clinical outcome in COVID-19 patients, including hospitalisation, ICU admission, mechanical ventilation and death, using registry data. Conclusion We identified FXR as a novel master regulator of ACE2 expression. Using a bench-to-bedside approach we combined in vitro, ex-vivo and patient data to demonstrate the efficacy of ACE2 downregulation against SARS-CoV-2 infection and identified approved and inexpensive drugs (UDCA, ZGG) which could be repurposed as prophylactic and therapeutic agents against SARS-CoV-2 infection, paving the road for future clinical trials.
- Published
- 2021
16. Author response: TGFβ signalling is required to maintain pluripotency of human naïve pluripotent stem cells
- Author
-
Ludovic Vallier, Brandon T Wesley, Peter J. Rugg-Gunn, Christel Krueger, Anna Osnato, A Sophie Brumm, Mariana Quiroga Londoño, Amanda J. Collier, Kathy K. Niakan, Stephanie Brown, Daniel Ortmann, Simon Andrews, Daniele Muraro, and Shota Nakanoh
- Subjects
Biology ,Induced pluripotent stem cell ,Tgfβ signalling ,Cell biology - Published
- 2021
17. Modeling PNPLA3-Associated NAFLD Using Human-Induced Pluripotent Stem Cells
- Author
-
An-Sofie Lenaerts, Samantha G. Tilson, Ludovic Vallier, Albert Koulman, T. Jake Liang, Zongyi Hu, Benjamin Jenkins, Seung Bum Park, and Carola M. Morell
- Subjects
Induced Pluripotent Stem Cells ,Biology ,Polymorphism, Single Nucleotide ,Cell Line ,Transcriptome ,Pathogenesis ,Gene Knockout Techniques ,Loss of Function Mutation ,Non-alcoholic Fatty Liver Disease ,medicine ,Toxicity Tests, Acute ,CRISPR ,Humans ,Genetic Predisposition to Disease ,Induced pluripotent stem cell ,Gene ,Loss function ,Hepatology ,Ethanol ,Membrane Proteins ,Cell Differentiation ,Lipase ,medicine.disease ,Lipid Metabolism ,Phenotype ,Cell biology ,Methotrexate ,Hepatocytes ,Steatosis ,CRISPR-Cas Systems - Abstract
BACKGROUND AND AIMS NAFLD is a growing public health burden. However, the pathogenesis of NAFLD has not yet been fully elucidated, and the importance of genetic factors has only recently been appreciated. Genomic studies have revealed a strong association between NAFLD progression and the I148M variant in patatin-like phospholipase domain-containing protein 3 (PNPLA3). Nonetheless, very little is known about the mechanisms by which this gene and its variants can influence disease development. To investigate these mechanisms, we have developed an in vitro model that takes advantage of the unique properties of human-induced pluripotent stem cells (hiPSCs) and the CRISPR/CAS9 gene editing technology. APPROACH AND RESULTS We used isogenic hiPSC lines with either a knockout (PNPLA3KO ) of the PNPLA3 gene or with the I148M variant (PNPLA3I148M ) to model PNPLA3-associated NAFLD. The resulting hiPSCs were differentiated into hepatocytes, treated with either unsaturated or saturated free fatty acids to induce NAFLD-like phenotypes, and characterized by various functional, transcriptomic, and lipidomic assays. PNPLA3KO hepatocytes showed higher lipid accumulation as well as an altered pattern of response to lipid-induced stress. Interestingly, loss of PNPLA3 also caused a reduction in xenobiotic metabolism and predisposed PNPLA3KO cells to be more susceptible to ethanol-induced and methotrexate-induced toxicity. The PNPLA3I148M cells exhibited an intermediate phenotype between the wild-type and PNPLA3KO cells. CONCLUSIONS Together, these results indicate that the I148M variant induces a loss of function predisposing to steatosis and increased susceptibility to hepatotoxins.
- Published
- 2021
18. FXR inhibition reduces ACE2 expression, SARS-CoV-2 infection and may improve COVID-19 outcome
- Author
-
Olivia C. Tysoe, Andrew M. Moon, Paul Gibbs, Sally Forrest, Sanjay Sinha, Johannes Bargehr, Daniele Muraro, Sidney A Barritt, William Gelson, Gwilym J. Webb, Eleanor Barnes, Mailis Maes, Stephen Baker, Gareth Corbett, Thomas L Williams, Gordon Dougan, Vasileios Galanakis, Ravindra K. Gupta, Fotios Sampaziotis, Scott Dillon, Ludovic Vallier, Lisa Swift, Teresa Brevini, Andrew J. Butler, Mahnaz Darvish-Damavandi, Sara Upponi, Ansgar W. Lohse, George F. Mells, Kourosh Saeb-Parsy, Simon J.A. Buczacki, Marta Vila-Gonzalez, Victoria L. Mulcahy, Susan E. Davies, Sagar Varankar, Thomas Marjot, Petra Mlcochova, Christopher J.E. Watson, Anthony P. Davenport, Joo-Hyeon Lee, Thomas W M Crozier, and Rhoda E. Kuc
- Subjects
Therapeutic approach ,Downregulation and upregulation ,business.industry ,Regulator ,Medicine ,Farnesoid X receptor ,Receptor ,business ,Bioinformatics ,Ursodeoxycholic acid ,Ex vivo ,Cholangiocyte ,medicine.drug - Abstract
Prevention of SARS-CoV-2 entry in cells through the modulation of viral host receptors, such as ACE2, could represent a new therapeutic approach complementing vaccination. However, the mechanisms controlling ACE2 expression remain elusive. Here, we identify the farnesoid X receptor (FXR) as a direct regulator of ACE2 transcription in multiple COVID19-affected tissues, including the gastrointestinal and respiratory systems. We demonstrate that FXR antagonists, including the over-the-counter compound z-guggulsterone (ZGG) and the off-patent drug ursodeoxycholic acid (UDCA), downregulate ACE2 levels, and reduce susceptibility to SARS-CoV-2 infection in lung, cholangiocyte and gut organoids. We then show that therapeutic levels of UDCA downregulate ACE2 in human organs perfused ex situ and reduce SARS-CoV-2 infection ex vivo. Finally, we perform a retrospective study using registry data and identify a correlation between UDCA treatment and positive clinical outcomes following SARS-CoV-2 infection, including hospitalisation, ICU admission and death. In conclusion, we identify a novel function of FXR in controlling ACE2 expression and provide evidence that this approach could be beneficial for reducing SARS-CoV-2 infection, thereby paving the road for future clinical trials.
- Published
- 2021
19. BS29 Single cell transcriptomics reveals regenerative embryonic pathways lost in the adult epicardium
- Author
-
Hongorzul Davaapil, Sanjay Sinha, Ludovic Vallier, Vincent Knight-Schrijver, Alexander Ross, and Laure Gambardella
- Subjects
Cell type ,biology ,business.industry ,Regeneration (biology) ,medicine.medical_treatment ,Cell ,Embryo ,Stem-cell therapy ,medicine.disease ,biology.organism_classification ,Embryonic stem cell ,Cell biology ,medicine.anatomical_structure ,Heart failure ,medicine ,business ,Zebrafish - Abstract
Background Damage to the heart muscle often leads to chronic arrhythmia and heart failure. This is because the muscle of the adult human heart does not functionally regenerate after injury and is replaced by fibrous non-contractile and non-conductive scar tissue. However, the heart muscle of some vertebrates and developing mammals is capable of regeneration which restores the muscular function of the damaged tissue. Evidence suggests that the epicardium plays a crucial role in orchestrating this successful regenerative response in animals like the zebrafish and that epicardium cells are active in developmental stages of life. However, in adult humans the epicardium is putatively quiescent. Objectives We hypothesise that the difference in the regenerative capacity of human adult and foetal myocardium may be explained in part by differences in the epicardium and reactivation of these molecular processes lost in adult epicardium may lead to effective myocardial regeneration. Using single-cell RNA sequencing, our aim was to capture the regenerative epicardial signalling present in the foetal epicardium that may be absent in adult hearts. Methods Heart sections from 7 human embryos between 8 and 12 weeks of gestation were dissociated and sequenced with scRNA-seq. ScRNA-seq data from 5 human adult hearts between 55 and 75 years old were acquired from the Heart Cell Atlas. Data were integrated and epicardial cells in all samples were identified using supervised classification and canonical markers. Results We compared the markers expressed in foetal and adult mesothelial epicardial cells and identified foetal, adult and stage-independent epicardial signatures. Among these were a number of pro-angiogenic factors specific to the foetal epicardium, including a group of WNT-signalling secreted factors, WNT2B, SFRP2 and SFRP5. Gene Ontology suggested a shift towards immune response of the epicardial cells during maturation. Additionally, an epicardial cluster with fibroblast-like markers was found only in the foetal heart (96 % foetal). We examined organ-wide influences of epicardial signalling by mapping the potential ligand receptor interactions from the epicardial-derived secreted factors to receptors upregulated in other heart cell clusters. This revealed the prospective influence of reactivating foetal epicardial pathways for heart regeneration. Lastly, we demonstrated that epicardium derived from human embryonic stem cells exhibits some of these lost regenerative pathways, promoting their value as a model and as therapeutic agents. Conclusions We have shown that the adult epicardium has major differences to the foetal epicardium. Firstly, a set of epicardium-specific angiogenic pathways are absent from the adult epicardium and secondly, foetal hearts contain another epicardial cell type that is not present in the adult heart. Recapitulating both the missing epicardial signals and the synergy between the foetal epicardial cell types may be essential for developing effective regenerative stem cell therapy. Conflict of Interest None
- Published
- 2021
20. Cell cycle regulators control mesoderm specification in human pluripotent stem cells
- Author
-
Rodrigo A. Grandy, Loukia Yiangou, Daniel Ortmann, Ludovic Vallier, Anna Osnato, Sanjay Sinha, Yiangou, Loukia [0000-0003-1477-005X], Vallier, Ludovic [0000-0002-3848-2602], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Mesoderm ,animal structures ,MAP Kinase Signaling System ,Ubiquitin-Protein Ligases ,education ,Human Embryonic Stem Cells ,tissue regeneration ,Germ layer ,Biology ,Biochemistry ,03 medical and health sciences ,stem cells ,Paraxial mesoderm ,medicine ,Humans ,Cell Lineage ,Induced pluripotent stem cell ,Protein Kinase Inhibitors ,Molecular Biology ,030102 biochemistry & molecular biology ,Cell Cycle ,Embryogenesis ,embryo development ,Cell Differentiation ,RB transcriptional corepressor 1 (RB1) ,differentiation ,Cell Biology ,cyclin-dependent kinase (CDK) ,pluripotency ,Cyclin-Dependent Kinases ,3. Good health ,Cell biology ,Gastrulation ,Retinoblastoma Binding Proteins ,030104 developmental biology ,medicine.anatomical_structure ,embryonic structures ,Mesoderm formation ,gene expression ,Stem cell ,signaling - Abstract
The mesoderm is one of the three germ layers produced during gastrulation from which muscle, bones, kidneys, and the cardiovascular system originate. Understanding the mechanisms that control mesoderm specification could inform many applications, including the development of regenerative medicine therapies to manage diseases affecting these tissues. Here, we used human pluripotent stem cells to investigate the role of cell cycle in mesoderm formation. To this end, using small molecules or conditional gene knockdown, we inhibited proteins controlling G1 and G2/M cell cycle phases during the differentiation of human pluripotent stem cells into lateral plate, cardiac, and presomitic mesoderm. These loss-of-function experiments revealed that regulators of the G1 phase, such as cyclin-dependent kinases and pRb (retinoblastoma protein), are necessary for efficient mesoderm formation in a context-dependent manner. Further investigations disclosed that inhibition of the G2/M regulator cyclin-dependent kinase 1 decreases BMP (bone morphogenetic protein) signaling activity specifically during lateral plate mesoderm formation while reducing fibroblast growth factor/extracellular signaling-regulated kinase 1/2 activity in all mesoderm subtypes. Taken together, our findings reveal that cell cycle regulators direct mesoderm formation by controlling the activity of key developmental pathways.
- Published
- 2019
21. Modeling Disease with Human Inducible Pluripotent Stem Cells
- Author
-
Ludovic Vallier, Rodrigo A. Grandy, Rute A. Tomaz, Vallier, Ludovic [0000-0002-3848-2602], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Computational biology ,Disease ,Biology ,liver ,Models, Biological ,Pathology and Forensic Medicine ,03 medical and health sciences ,Liver disease ,0302 clinical medicine ,disease modeling ,medicine ,Humans ,Human Induced Pluripotent Stem Cells ,Induced pluripotent stem cell ,hiPSCs ,reprogramming ,Cell Differentiation ,differentiation ,medicine.disease ,Model disease ,3. Good health ,030104 developmental biology ,Hepatocytes ,Reprogramming ,030217 neurology & neurosurgery - Abstract
Understanding the physiopathology of disease remains an essential step in developing novel therapeutics. Although animal models have certainly contributed to advancing this enterprise, their limitation in modeling all the aspects of complex human disorders is one of the major challenges faced by the biomedical research field. Human induced pluripotent stem cells (hiPSCs) derived from patients represent a great opportunity to overcome this deficiency because these cells cover the genetic diversity needed to fully model human diseases. Here, we provide an overview of the history of hiPSC technology and discuss common challenges and approaches that we and others have faced when using hiPSCs to model disease. Our emphasis is on liver disease, and consequently, we review the progress made using this technology to produce functional liver cells in vitro and how these systems are being used to recapitulate a diversity of developmental, metabolic, genetic, and infectious liver disorders.
- Published
- 2019
22. Chronic injury induces plasticity between cholangiocytes and hepatocytes in the human liver
- Author
-
Vasileios Galanakis, Christopher Gribben, Alexander Calderwood, Ilias Moutsopoulos, Katarzyna Kania, Emmanouil Athanasiadis, Michael Allison, Irina Mohorianu, and Ludovic Vallier
- Subjects
Hepatology - Published
- 2022
23. Science-based assessment of source materials for cell-based medicines: report of a stakeholders workshop
- Author
-
Louise Bisset, David Pan, Julian Braybrook, Raul Elgueta Rebolledo, Marcelo N. Rivolta, Christopher E. Goldring, Sujith Sebastian, B. Kevin Park, Angela E Thomas, Eric J. Roos, John Gardner, Bernd Schröder, Roland Leathers, Jacqueline Barry, Andrew Webster, Ludovic Vallier, Michael H Neale, Glyn Stacey, Susan J. Kimber, Peter W. Andrews, Philip Driver, Sharon Patricia Mary Crouch, Loriana Vitillo, Robin Buckle, Helen Jesson, Lyn Healy, Peter J. Coffey, David J. Williams, Anna Hows, Amit Chandra, Claire Hutchinson, Robert Thomas, Sarah Moyle, Allan Ritchie, Ivana Barbaric, Marc Turner, Trish Murray, Tammy L. Kalber, David C. Hay, Amanda L. Evans, Curtis O. Asante, Kourosh Saeb-Parsy, Patrick J. Ginty, and Ian Rees
- Subjects
Pluripotent Stem Cells ,safety ,0301 basic medicine ,Embryology ,efficacy ,starting materials ,Cell- and Tissue-Based Therapy ,Biomedical Engineering ,regenerative medicine ,Regenerative Medicine ,Key issues ,03 medical and health sciences ,0302 clinical medicine ,cell-based medicines ,Biomanufacturing ,characterization regulatory science ,United Kingdom ,030104 developmental biology ,Practice Guidelines as Topic ,raw materials ,Engineering ethics ,Patient Safety ,cell therapy ,ancillary materials ,030217 neurology & neurosurgery ,Cell based - Abstract
Human pluripotent stem cells (hPSCs) have the potential to transform medicine. However, hurdles remain to ensure safety for such cellular products. Science-based understanding of the requirements for source materials is required as are appropriate materials. Leaders in hPSC biology, clinical translation, biomanufacturing and regulatory issues were brought together to define requirements for source materials for the production of hPSC-derived therapies and to identify other key issues for the safety of cell therapy products. While the focus of this meeting was on hPSC-derived cell therapies, many of the issues are generic to all cell-based medicines. The intent of this report is to summarize the key issues discussed and record the consensus reached on each of these by the expert delegates.
- Published
- 2018
24. Modeling HNF1B-associated monogenic diabetes using human iPSCs reveals an early stage impairment of the pancreatic developmental program
- Author
-
Ludovic Vallier, Pedro Madrigal, Daniele Muraro, Katarzyna Tilgner, Mariya Chhatriwala, Sapna Vyas, Crystal Y. Chia, Evelyn Olszanowski, Ranna El-Khairi, and Santiago A. Rodríguez-Seguí
- Subjects
HNF1B ,Organogenesis ,Induced Pluripotent Stem Cells ,Fluorescent Antibody Technique ,β cell ,Haploinsufficiency ,Biology ,Biochemistry ,Models, Biological ,Article ,Immunophenotyping ,Downregulation and upregulation ,Insulin-Secreting Cells ,monogenic ,Genetics ,medicine ,Diabetes Mellitus ,Humans ,Cell Lineage ,pancreas ,Progenitor cell ,Induced pluripotent stem cell ,TEAD1 ,in vitro ,Progenitor ,Hepatocyte Nuclear Factor 1-beta ,Gene Editing ,iPSC ,diabetes ,Gene Expression Profiling ,MODY5 ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell Biology ,differentiation ,human induced pluripotent stem cells ,medicine.anatomical_structure ,Cancer research ,Disease Susceptibility ,CRISPR-Cas Systems ,Pancreas ,Biomarkers ,Developmental Biology ,Signal Transduction - Abstract
Summary Heterozygous mutations in HNF1B in humans result in a multisystem disorder, including pancreatic hypoplasia and diabetes mellitus. Here we used a well-controlled human induced pluripotent stem cell pancreatic differentiation model to elucidate the molecular mechanisms underlying HNF1B-associated diabetes. Our results show that lack of HNF1B blocks specification of pancreatic fate from the foregut progenitor (FP) stage, but HNF1B haploinsufficiency allows differentiation of multipotent pancreatic progenitor cells (MPCs) and insulin-secreting β-like cells. We show that HNF1B haploinsufficiency impairs cell proliferation in FPs and MPCs. This could be attributed to impaired induction of key pancreatic developmental genes, including SOX11, ROBO2, and additional TEAD1 target genes whose function is associated with MPC self-renewal. In this work we uncover an exhaustive list of potential HNF1B gene targets during human pancreas organogenesis whose downregulation might underlie HNF1B-associated diabetes onset in humans, thus providing an important resource to understand the pathogenesis of this disease., Graphical abstract, Highlights • Lack of HNF1B blocks specification of pancreatic fate from the FP stage • HNF1B haploinsufficiency allows differentiation of MPCs and β-like cells • HNF1B haploinsufficiency impairs cell proliferation in FPs and MPCs • Reduced HNF1B levels impair the induction of SOX11, ROBO2, and TEAD1 targets, In this article, Rodríguez-Seguí and colleagues use an hiPSC pancreatic differentiation model to study the mechanisms underlying HNF1B-associated diabetes. The authors show that lack of HNF1B blocks specification of pancreatic fate from the foregut progenitor (FP) stage. HNF1B haploinsufficiency, on the other hand, allows differentiation of multipotent pancreatic progenitor cells (MPCs) and insulin-secreting β-like cells, but impairs cell proliferation in FPs and MPCs.
- Published
- 2021
25. Organoids capture tissue-specific innate lymphoid cell development in mice and humans
- Author
-
Geraldine M. Jowett, Emily Read, Luke B. Roberts, Diana Coman, Marta Vilà González, Tomasz Zabinski, Umar Niazi, Rita Reis, Tung-Jui Trieu, Davide Danovi, Eileen Gentleman, Ludovic Vallier, Michael A. Curtis, Graham M. Lord, and Joana F. Neves
- Subjects
Organoids ,Mice ,Induced Pluripotent Stem Cells ,Animals ,Humans ,Cell Differentiation ,Immunotherapy ,Lymphocytes ,General Biochemistry, Genetics and Molecular Biology ,Immunity, Innate - Abstract
Organoid-based models of murine and human innate lymphoid cell precursor (ILCP) maturation are presented. First, murine intestinal and pulmonary organoids are harnessed to demonstrate that the epithelial niche is sufficient to drive tissue-specific maturation of all innate lymphoid cell (ILC) groups in parallel, without requiring subset-specific cytokine supplementation. Then, more complex human induced pluripotent stem cell (hiPSC)-based gut and lung organoid models are used to demonstrate that human epithelial cells recapitulate maturation of ILC from a stringent systemic human ILCP population, but only when the organoid-associated stromal cells are depleted. These systems offer versatile and reductionist models to dissect the impact of environmental and mucosal niche cues on ILC maturation. In the future, these could provide insight into how ILC activity and development might become dysregulated in chronic inflammatory diseases.
- Published
- 2021
26. Author response: An in vitro stem cell model of human epiblast and yolk sac interaction
- Author
-
Marta N. Shahbazi, Tim H. H. Coorens, Charlotte E Handford, Bailey A. T. Weatherbee, Lygia da Veiga Pereira, Sam Behjati, George Hudson, Magdalena Zernicka-Goetz, Ludovic Vallier, Kirsty Ml Mackinlay, and Viviane Souza Rosa
- Subjects
medicine.anatomical_structure ,Epiblast ,medicine ,Yolk sac ,Biology ,Stem cell ,In vitro ,Cell biology - Published
- 2021
27. TGFβ signalling is required to maintain pluripotency of human naïve pluripotent stem cells
- Author
-
Ludovic Vallier, Peter J. Rugg-Gunn, Simon Andrews, Daniel Ortmann, Stephanie Brown, A Sophie Brumm, Anna Osnato, Amanda J. Collier, Daniele Muraro, Christel Krueger, Kathy K. Niakan, Shota Nakanoh, Brandon T Wesley, Mariana Quiroga Londoño, Osnato, Anna [0000-0001-5241-1512], Krueger, Christel [0000-0001-5601-598X], Andrews, Simon [0000-0002-5006-3507], Collier, Amanda J [0000-0003-1137-6874], Quiroga Londoño, Mariana [0000-0003-2352-0773], Niakan, Kathy K [0000-0003-1646-4734], Vallier, Ludovic [0000-0002-3848-2602], Rugg-Gunn, Peter J [0000-0002-9601-5949], and Apollo - University of Cambridge Repository
- Subjects
Pluripotent Stem Cells ,QH301-705.5 ,Science ,Smad2 Protein ,Biology ,Regenerative Medicine ,Regenerative medicine ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Transforming Growth Factor beta ,Humans ,Smad3 Protein ,Biology (General) ,Beta (finance) ,Induced pluripotent stem cell ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,General Immunology and Microbiology ,Neuroectoderm ,Effector ,General Neuroscience ,Stem Cells ,Cell Differentiation ,General Medicine ,embryonic stem cells ,Cellular Reprogramming ,Embryonic stem cell ,Stem Cells and Regenerative Medicine ,3. Good health ,Cell biology ,Regulatory sequence ,Medicine ,Stem cell ,NODAL ,gene regulation ,030217 neurology & neurosurgery ,Transforming growth factor ,Signal Transduction ,Research Article ,Human - Abstract
The signalling pathways that maintain primed human pluripotent stem cells (hPSCs) have been well characterised, revealing a critical role for TGFβ/Activin/Nodal signalling. In contrast, the signalling requirements of naïve human pluripotency have not been fully established. Here, we demonstrate that TGFβ signalling is required to maintain naïve hPSCs. The downstream effector proteins – SMAD2/3 – bind common sites in naïve and primed hPSCs, including shared pluripotency genes. In naïve hPSCs, SMAD2/3 additionally bind to active regulatory regions near to naïve pluripotency genes. Inhibiting TGFβ signalling in naïve hPSCs causes the downregulation of SMAD2/3-target genes and pluripotency exit. Single-cell analyses reveal that naïve and primed hPSCs follow different transcriptional trajectories after inhibition of TGFβ signalling. Primed hPSCs differentiate into neuroectoderm cells, whereas naïve hPSCs transition into trophectoderm. These results establish that there is a continuum for TGFβ pathway function in human pluripotency spanning a developmental window from naïve to primed states.
- Published
- 2021
28. Building consensus on definition and nomenclature of hepatic, pancreatic, and biliary organoids
- Author
-
Ary Marsee, Floris J.M. Roos, Monique M.A. Verstegen, Helmuth Gehart, Eelco de Koning, Frédéric Lemaigre, Stuart J. Forbes, Weng Chuan Peng, Meritxell Huch, Takanori Takebe, Ludovic Vallier, Hans Clevers, Luc J.W. van der Laan, Bart Spee, Floris Roos, Monique Verstegen, Stuart Forbes, Luc van der Laan, Sylvia Boj, Pedro Baptista, Kerstin Schneeberger, Carol Soroka, Markus Heim, Sandro Nuciforo, Kenneth Zaret, Yoshimasa Saito, Matthias Lutolf, Vincenzo Cardinale, Ben Simons, Sven van IJzendoorn, Akihide Kamiya, Hiromi Chikada, Shuyong Wang, Seon Ju Mun, Myung Jin Son, Tamer Tevfik Onder, James Boyer, Toshiro Sato, Nikitas Georgakopoulos, Andre Meneses, Laura Broutier, Luke Boulter, Dominic Grün, Jan IJzermans, Benedetta Artegiani, Ruben van Boxtel, Ewart Kuijk, Guido Carpino, Gary Peltz, Jesus Banales, Nancy Man, Luigi Aloia, Nicholas LaRusso, Gregory George, Casey Rimland, George Yeoh, Anne Grappin-Botton, Daniel Stange, Nicole Prior, Janina E.E. Tirnitz-Parker, Emma Andersson, Chiara Braconi, Nicholas Hannan, Wei-Yu Lu, Stephen Strom, Pau Sancho-Bru, Shinichiro Ogawa, Vincenzo Corbo, Madeline Lancaster, Huili Hu, Sabine Fuchs, Delilah Hendriks, Roos, Floris Johan Maria [0000-0003-1278-6517], Apollo - University of Cambridge Repository, Surgery, and UCL - SSS/DDUV/LPAD - Liver and pancreas differentiation
- Subjects
Consensus ,Standardization ,Delphi method ,tumor organoid ,Biology ,liver ,03 medical and health sciences ,0302 clinical medicine ,bile duct ,Organoid ,Genetics ,pancreas ,gallbladder ,030304 developmental biology ,0303 health sciences ,multi-organ organoid ,Management science ,epithelial organoid ,Cell Biology ,multi-tissue organoid ,3. Good health ,Organoids ,Mouse Pancreas ,HPB ,Molecular Medicine ,Adult liver ,030217 neurology & neurosurgery - Abstract
Hepatic, pancreatic, and biliary (HPB) organoids are powerful tools for studying development, disease, and regeneration. As organoid research expands, the need for clear definitions and nomenclature describing these systems also grows. To facilitate scientific communication and consistent interpretation, we revisit the concept of an organoid and introduce an intuitive classification system and nomenclature for describing these 3D structures through the consensus of experts in the field. To promote the standardization and validation of HPB organoids, we propose guidelines for establishing, characterizing, and benchmarking future systems. Finally, we address some of the major challenges to the clinical application of organoids.
- Published
- 2021
- Full Text
- View/download PDF
29. Derivation of Multipotent Neural Progenitors from Human Embryonic Stem Cells for Cell Therapy and Biomedical Applications
- Author
-
Ludovic Vallier, Loriana Vitillo, Vallier, Ludovic [0000-0002-3848-2602], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Neurogenesis ,viruses ,Human Embryonic Stem Cells ,Cell ,Population ,Cell- and Tissue-Based Therapy ,Stem cells ,Biology ,environment and public health ,Regenerative medicine ,Cell therapy ,03 medical and health sciences ,Neural Stem Cells ,medicine ,Humans ,Progenitor cell ,education ,Lt-NES ,Progenitor ,education.field_of_study ,030102 biochemistry & molecular biology ,GMP ,Cell Differentiation ,Neural progenitors ,Embryonic stem cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Differentiation ,lipids (amino acids, peptides, and proteins) ,Stem cell - Abstract
Long-term neuroepithelial-like stem cells (lt-NES) derived from human embryonic stem cells are a stable self-renewing progenitor population with high neurogenic potential and phenotypic plasticity. Lt-NES are amenable to regional patterning toward neurons and glia subtypes and thus represent a valuable source of cells for many biomedical applications. For use in regenerative medicine and cell therapy, lt-NES and their progeny require derivation with high-quality culture conditions suitable for clinical use. In this chapter, we describe a robust method to derive multipotent and expandable lt-NES based on good manufacturing practice and cell therapy-grade reagents. We further describe fully defined protocols to terminally differentiate lt-NES toward GABA-ergic, dopaminergic, and motor neurons.
- Published
- 2021
30. Naive Pluripotent Stem Cells Exhibit Phenotypic Variability that Is Driven by Genetic Variation
- Author
-
Daniel Ortmann, Daniele Muraro, Laura G. Reinholdt, Brandon T Wesley, Ludovic Vallier, Daniel A. Skelly, Christopher L. Baker, Ted Choi, Yuanhua Huang, Peter J. Rugg-Gunn, Rute A. Tomaz, Steven C. Munger, Stephanie Brown, Anna Osnato, Gary A. Churchill, Anne Czechanski, Selcan Aydin, Oliver Stegle, Giovanni Canu, Osnato, Anna [0000-0001-5241-1512], Wesley, Brandon [0000-0003-0530-329X], Vallier, Ludovic [0000-0002-3848-2602], and Apollo - University of Cambridge Repository
- Subjects
Pluripotent Stem Cells ,education ,Cell ,Induced Pluripotent Stem Cells ,mouse embryonic stem cells ,naïve ,Biology ,eQTL ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Short Article ,Genetic variation ,medicine ,Animals ,genetics ,ground state ,Epigenetics ,Induced pluripotent stem cell ,health care economics and organizations ,030304 developmental biology ,Genetics ,0303 health sciences ,variability ,Wnt signaling pathway ,Genetic Variation ,Reproducibility of Results ,Cell Differentiation ,Cell Biology ,differentiation ,Phenotype ,medicine.anatomical_structure ,Biological Variation, Population ,Expression quantitative trait loci ,Molecular Medicine ,Stem cell ,signaling ,030217 neurology & neurosurgery - Abstract
Summary Variability among pluripotent stem cell (PSC) lines is a prevailing issue that hampers not only experimental reproducibility but also large-scale applications and personalized cell-based therapy. This variability could result from epigenetic and genetic factors that influence stem cell behavior. Naive culture conditions minimize epigenetic fluctuation, potentially overcoming differences in PSC line differentiation potential. Here we derived PSCs from distinct mouse strains under naive conditions and show that lines from distinct genetic backgrounds have divergent differentiation capacity, confirming a major role for genetics in PSC phenotypic variability. This is explained in part through inconsistent activity of extra-cellular signaling, including the Wnt pathway, which is modulated by specific genetic variants. Overall, this study shows that genetic background plays a dominant role in driving phenotypic variability of PSCs., Graphical Abstract, Highlights • Ground-state mESCs are variable in gene expression and capacity of differentiation • Genetic background contributes to ESC variability even after epigenetic resetting • Signaling pathway activity is influenced by genetic traits • Analysis of eQTL identifies genetic elements causing variability, Ortmann et al. show that ground-state pluripotent stem cells exhibit variability in gene expression and differentiation propensity that is associated with their genetic background. This variability is linked to differences in major signaling pathway activity, showing that analyses of expression quantitative trait loci enable identification of causal genetic elements.
- Published
- 2020
- Full Text
- View/download PDF
31. A spatially resolved single cell atlas of human gastrulation
- Author
-
Antonio Scialdone, Elmir Mahammadov, Shankar Srinivas, Richard C. V. Tyser, Ludovic Vallier, and Shota Nakanoh
- Subjects
Gastrulation ,Transcriptome ,Body plan ,Directed differentiation ,Epiblast ,Embryogenesis ,Embryo ,Biology ,Embryonic stem cell ,Cell biology - Abstract
Gastrulation is the fundamental process during the embryogenesis of all multicellular animals through which the basic body plan is first laid down. It is pivotal in generating cellular diversity coordinated with spatial patterning. Gastrulation in humans occurs in the third week following fertilization. Our understanding of this process in humans is extremely limited, and based almost entirely on experimental models. Here, we characterize in a spatially resolved manner the single cell transcriptional profile of an entire gastrulating human embryo approximately 16 to 19 days after fertilization. We used these data to provide the first unequivocal demonstration that human embryonic stem cells represent the early post implantation epiblast. We identified both primordial germ cells and red blood cells, which had never been characterized so early during human development. Comparison with mouse gastrula transcriptomes revealed many commonalities between the human and mouse but also several key differences, particularly in FGF signaling, that we validated experimentally. This unique dataset offers a unique glimpse into a central but generally inaccessible stage of our development, provides new context for interpreting experiments in other model systems and represents a valuable resource for guiding directed differentiation of human cellsin vitro.
- Published
- 2020
32. Unravelling the developmental roadmap towards human brown adipose tissue
- Author
-
Barry Rosen, Ludovic Vallier, Davide Chiarugi, Sasha Mendjan, Isabella Samuelson, Myriam Bahri, Kathleen Long, Dunja Lukovic, Andrew R. Bassett, Floris Honig, Sherine Awad, Anne-Claire Guenantin, Stefania Carobbio, Ioannis Kamzolas, Antonio Vidal-Puig, Slaven Erceg, and Sonia Rodríguez-Fdez
- Subjects
animal structures ,Brown Adipocytes ,Biology ,Low volume ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Adipocyte ,Brown adipose tissue ,medicine ,Paraxial mesoderm ,Induced pluripotent stem cell ,Neuroscience ,Therapeutic strategy ,Progenitor - Abstract
Increasing brown adipose tissue (BAT) mass and activation has been proposed as a potential therapeutic strategy to treat obesity and associated cardiometabolic complications. Given that obese and diabetic patients possess low amounts of BAT, an efficient way to expand their BAT mass would be necessary if BAT is to be useful. Currently, there is limited knowledge about how human BAT develops, differentiates, and is optimally activated. Moreover, to have access to human BAT is challenging, given its low volume and being anatomically dispersed. These constrain makes detailed mechanistic studies related to BAT development and function in humans virtually impossible. To overcome these limitations, we have developed a human-relevant new protocol for the differentiation of human pluripotent stem cells (hPSCs) into brown adipocytes (BAs). Unique to this protocol is that it is chemically-defined to recapitulate a physiological step-by-step developmental path of BAT that captures transient paraxial mesoderm and BAT progenitor states, on its way to reaching the adipocyte stage finally. These hPSC-derived BAs express brown adipocyte and thermogenic markers, are insulin sensitive, and respond to β-adrenergic stimuli. This new protocol is a scalable tool to study human BAs during development.
- Published
- 2020
33. Epigenetic regulations follow cell cycle progression during differentiation of human pluripotent stem cells
- Author
-
Rodrigo A. Grandy, Anna Osnato, Pedro Madrigal, Siim Pauklin, Daniel Ortmann, Ludovic Vallier, Kim Jee Goh, and Stephanie Brown
- Subjects
medicine.anatomical_structure ,Cell division ,medicine ,Germ layer ,Cell cycle ,Stem cell ,Endoderm ,Cell fate determination ,Biology ,Induced pluripotent stem cell ,Embryonic stem cell ,Cell biology - Abstract
Most mammalian stem cells undergo cellular division during their differentiation to produce daughter cells with a new cellular identity. However, the cascade of epigenetic events and molecular mechanisms occurring between successive cell divisions upon differentiation have not yet been described in detail due to technical limitations. Here, we address this question by taking advantage of the Fluorescent Ubiquitination-based Cell Cycle Indicator (FUCCI) reporter to develop a culture system allowing the differentiation of human Embryonic Stem Cells (hESCs) synchronised for their cell cycle. Using this approach, we have assessed the epigenome and transcriptome dynamics during the first two divisions leading to definitive endoderm. We first observed that transcription of key markers of differentiation occurs before division suggesting that differentiation is initiated during the progression of cell cycle. Furthermore, ATAC-seq shows a major decrease in chromatin accessibility after pluripotency exit indicating that the first event of differentiation is the inhibition of alternative cell fate. In addition, using digital genomic footprinting we identified novel cell cycle-specific transcription factors with regulatory potential in endoderm specification. Of particular interest, Activator protein 1 (AP-1) controlled p38/MAPK signalling seems to be necessary for blocking endoderm shifting cell fate toward mesoderm lineage. Finally, histone modifications analyses suggest a temporal order between different marks. We can also conclude that enhancers are dynamically and rapidly established / decommissioned between different cell cycle upon differentiation. Overall, these data not only reveal key the successive interplays between epigenetic modifications during differentiation but also provide a valuable resource to investigate novel mechanisms in germ layer specification.
- Published
- 2020
34. Transcriptional networks are dynamically regulated during cell cycle progression in human Pluripotent Stem Cells
- Author
-
Anna Osnato, Stephanie Brown, and Ludovic Vallier
- Subjects
Homeobox protein NANOG ,SOX2 ,CTCF ,Cell cycle ,Biology ,Stem cell ,Induced pluripotent stem cell ,Embryonic stem cell ,Chromatin ,Cell biology - Abstract
SummaryCell cycle progression follows a precise sequence of events marked by different phases and checkpoints which are associated with specific chromatin organisation. Whilst these changes have been extensively studied, their consequences on transcriptional networks remain to be fully uncovered, especially in dynamic model systems such as stem cells. Here, we take advantage of the FUCCI reporter system to show that chromatin accessibility, gene expression and key transcription factors binding change during cell cycle progression in human Embryonic Stem Cells (hESCs). These analyses reveal that core pluripotency factors such as OCT4, NANOG and SOX2 but also chromatin remodelers such as CTCF and RING1B bind the genome at specific phases of the cell cycle. Importantly, this binding pattern allows differentiation in the G1 phase while preserving pluripotency in the S/G2/M. Our results highlight the importance of studying transcriptional and epigenetic regulations in the dynamic context of the cell cycle.
- Published
- 2020
35. Publisher Correction: Single-cell RNA-sequencing of differentiating iPS cells reveals dynamic genetic effects on gene expression
- Author
-
Anna S E Cuomo, Iker Martinez, Ludovic Vallier, Pedro Madrigal, Oliver Stegle, Andrew J Knights, Kedar Nath Natarajan, Shradha Amatya, Florian Buettner, Mariya Chhatriwala, John C. Marioni, Daniel D Seaton, Marc Jan Bonder, Abigail Isaacson, Davis J. McCarthy, and Jose Garcia-Bernardo
- Subjects
Male ,Quantitative trait loci ,Science ,Induced Pluripotent Stem Cells ,Cell ,Gene Expression ,General Physics and Astronomy ,Computational biology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Genetic Heterogeneity ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Gene expression ,medicine ,Humans ,lcsh:Science ,Induced pluripotent stem cell ,Genetic Association Studies ,Genetic association study ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,business.industry ,Gene Expression Profiling ,Endoderm ,RNA ,Functional genomics ,Cell Differentiation ,General Chemistry ,Publisher Correction ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Gene-Environment Interaction ,lcsh:Q ,Single-Cell Analysis ,business - Abstract
Recent developments in stem cell biology have enabled the study of cell fate decisions in early human development that are impossible to study in vivo. However, understanding how development varies across individuals and, in particular, the influence of common genetic variants during this process has not been characterised. Here, we exploit human iPS cell lines from 125 donors, a pooled experimental design, and single-cell RNA-sequencing to study population variation of endoderm differentiation. We identify molecular markers that are predictive of differentiation efficiency of individual lines, and utilise heterogeneity in the genetic background across individuals to map hundreds of expression quantitative trait loci that influence expression dynamically during differentiation and across cellular contexts.
- Published
- 2020
36. Single-cell RNA-sequencing of differentiating iPS cells reveals dynamic genetic effects on gene expression
- Author
-
Marc Jan Bonder, Oliver Stegle, Andrew J Knights, Davis J. McCarthy, Abigail Isaacson, Kedar Nath Natarajan, John C. Marioni, Daniel D Seaton, Jose Garcia-Bernardo, Mariya Chhatriwala, Shradha Amatya, Anna S E Cuomo, Pedro Madrigal, Iker Martinez, Ludovic Vallier, Florian Buettner, Seaton, Daniel D [0000-0002-5222-3893], McCarthy, Davis J [0000-0002-2218-6833], Madrigal, Pedro [0000-0003-1959-8199], Knights, Andrew [0000-0003-2107-4175], Natarajan, Kedar Nath [0000-0002-9264-1280], Vallier, Ludovic [0000-0002-3848-2602], Marioni, John C [0000-0001-9092-0852], Stegle, Oliver [0000-0002-8818-7193], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Male ,Science ,Induced Pluripotent Stem Cells ,Quantitative Trait Loci ,Cell ,General Physics and Astronomy ,Gene Expression ,Computational biology ,Quantitative trait locus ,Cell fate determination ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,Genetic Heterogeneity ,03 medical and health sciences ,0302 clinical medicine ,Single-cell analysis ,Gene expression ,medicine ,Humans ,lcsh:Science ,Induced pluripotent stem cell ,Genetic Association Studies ,Genetic association study ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Genetic heterogeneity ,Gene Expression Profiling ,Endoderm ,RNA ,Functional genomics ,Cell Differentiation ,General Chemistry ,Cell biology ,Gene expression profiling ,030104 developmental biology ,medicine.anatomical_structure ,Expression quantitative trait loci ,lcsh:Q ,Female ,Gene-Environment Interaction ,Single-Cell Analysis ,030217 neurology & neurosurgery ,Definitive endoderm - Abstract
Recent developments in stem cell biology have enabled the study of cell fate decisions in early human development that are impossible to study in vivo. However, understanding how development varies across individuals and, in particular, the influence of common genetic variants during this process has not been characterised. Here, we exploit human iPS cell lines from 125 donors, a pooled experimental design, and single-cell RNA-sequencing to study population variation of endoderm differentiation. We identify molecular markers that are predictive of differentiation efficiency of individual lines, and utilise heterogeneity in the genetic background across individuals to map hundreds of expression quantitative trait loci that influence expression dynamically during differentiation and across cellular contexts., Studying the genetic effects on early stages of human development is challenging due to a scarcity of biological material. Here, the authors utilise induced pluripotent stem cells from 125 donors to track gene expression changes and expression quantitative trait loci at single cell resolution during in vitro endoderm differentiation.
- Published
- 2020
- Full Text
- View/download PDF
37. Regional differences in human biliary tissues and corresponding in vitro derived organoids
- Author
-
Matthias Zilbauer, Olivia C. Tysoe, Daniele Muraro, Ludovic Vallier, Carola M. Morell, Nicholas R.F. Hannan, Stuart J. Forbes, Samantha G. Tilson, Richard L. Gieseck, Thomas A. Wynn, Simone E. Adams, Rute A. Tomaz, Brandon T Wesley, Wei-Yu Lu, Judith Kraiczy, John R. Ferdinand, Kourosh Saeb-Parsy, Gabriel C. Oniscu, Alexander Ross, Casey A. Rimland, Francisco Otaizo-Carrasquero, Nikitas Georgakopoulos, Timothy G. Myers, and Fotios Sampaziotis
- Subjects
0301 basic medicine ,Tissue and Organ Procurement ,Cellular differentiation ,Intrahepatic bile ducts ,Biology ,Cholangiocyte ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Liver Injury and Regeneration ,Bile Ducts, Extrahepatic ,Organoid ,medicine ,Animals ,Bile ,Humans ,RNA-Seq ,Pancreatic duct ,Common Bile Duct ,Keratin-19 ,Common bile duct ,Hepatology ,Gallbladder ,Wnt signaling pathway ,Cell Differentiation ,Epithelial Cells ,Original Articles ,Cell biology ,Organoids ,030104 developmental biology ,medicine.anatomical_structure ,Bile Ducts, Intrahepatic ,Gene Expression Regulation ,Liver ,030211 gastroenterology & hepatology ,Original Article - Abstract
Background and aims Organoids provide a powerful system to study epithelia in vitro. Recently, this approach was applied successfully to the biliary tree, a series of ductular tissues responsible for the drainage of bile and pancreatic secretions. More precisely, organoids have been derived from ductal tissue located outside (extrahepatic bile ducts; EHBDs) or inside the liver (intrahepatic bile ducts; IHBDs). These organoids share many characteristics, including expression of cholangiocyte markers such as keratin (KRT) 19. However, the relationship between these organoids and their tissues of origin, and to each other, is largely unknown. Approach and results Organoids were derived from human gallbladder, common bile duct, pancreatic duct, and IHBDs using culture conditions promoting WNT signaling. The resulting IHBD and EHBD organoids expressed stem/progenitor markers leucine-rich repeat-containing G-protein-coupled receptor 5/prominin 1 and ductal markers KRT19/KRT7. However, RNA sequencing revealed that organoids conserve only a limited number of regional-specific markers corresponding to their location of origin. Of particular interest, down-regulation of biliary markers and up-regulation of cell-cycle genes were observed in organoids. IHBD and EHBD organoids diverged in their response to WNT signaling, and only IHBDs were able to express a low level of hepatocyte markers under differentiation conditions. Conclusions Taken together, our results demonstrate that differences exist not only between extrahepatic biliary organoids and their tissue of origin, but also between IHBD and EHBD organoids. This information may help to understand the tissue specificity of cholangiopathies and also to identify targets for therapeutic development.
- Published
- 2020
- Full Text
- View/download PDF
38. Proteomic Comparison of Various Hepatic Cell Cultures for Preclinical Safety Pharmacology
- Author
-
Tracey Hurrell, Ludovic Vallier, Charis-Patricia Segeritz, A.D. Cromarty, and Kathryn S. Lilley
- Subjects
Proteomics ,0301 basic medicine ,Proteome ,Cellular differentiation ,Cell Culture Techniques ,Drug Evaluation, Preclinical ,Biology ,Toxicology ,Mass Spectrometry ,03 medical and health sciences ,0302 clinical medicine ,Spheroids, Cellular ,Humans ,Induced pluripotent stem cell ,Principal Component Analysis ,Solid Phase Extraction ,In vitro toxicology ,Cell Differentiation ,Hep G2 Cells ,Cell biology ,030104 developmental biology ,Cell culture ,Cancer cell ,Hepatocytes ,Hepatic stellate cell ,Electrophoresis, Polyacrylamide Gel ,030217 neurology & neurosurgery - Abstract
Experimental drugs need to be screened for safety within time constraints. Hepatotoxicity is one concerning contributor to the failure of investigational new drugs and a major rationale for postmarketing withdrawal decisions. Ethical considerations in preclinical research force the requirement for highly predictive in vitro assays using human tissue which retains functionality reflective of primary tissue. Here, the proteome of cells commonly used to assess preclinical hepatotoxicity was compared. Primary human hepatocytes (PHHs), hepatocyte-like cells (HLCs) differentiated from human pluripotent stem cells, HepG2 cell monolayers and HepG2 cell 3D spheroids were cultured and collected as whole cell lysates. Over 6000 proteins were identified and quantified in terms of relative abundance in replicate proteomic experiments using isobaric tagging methods. Comparison of these quantitative data provides biological insight into the feasibility of using HLCs, HepG2 monolayers, and HepG2 3D spheroids for hepatotoxicity testing. Collectively these data reveal how HLCs differentiated for 35 days and HepG2 cells proteomes differ from one another and that of PHHs. HepG2 cells possess a strong cancer cell signature and do not adequately express key metabolic proteins which mark the hepatic phenotype, this was not substantially altered by culturing as 3D spheroids. These data suggest that while no single hepatic model reflects the diverse array of outcomes required to mimic the in vivo liver functions, that HLCs are the most suitable investigational avenue for replacing PHHs in vitro.
- Published
- 2018
39. Advances in the generation of bioengineered bile ducts
- Author
-
Athina E. Markaki, Fotios Sampaziotis, Kourosh Saeb-Parsy, Alexander W. Justin, Ludovic Vallier, Saeb-Parsy, Kourosh [0000-0002-0633-3696], Markaki, Athina [0000-0002-2265-1256], Vallier, Ludovic [0000-0002-3848-2602], Sampaziotis, Fotios [0000-0003-0812-7586], and Apollo - University of Cambridge Repository
- Subjects
Cholangiopathy ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,Biomedical Engineering ,Cell Culture Techniques ,Bioengineering ,Bile Duct Diseases ,Liver transplantation ,Bioinformatics ,Cholangiocyte ,03 medical and health sciences ,Organ Culture Techniques ,Tissue engineering ,Biliary atresia ,Animals ,Humans ,Medicine ,Molecular Biology ,Bioartificial Organs ,Tissue Engineering ,business.industry ,Bile duct ,Epithelial Cells ,medicine.disease ,Coculture Techniques ,Liver Transplantation ,3. Good health ,Transplantation ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,Molecular Medicine ,Bile Ducts ,business - Abstract
The generation of bioengineered biliary tissue could contribute to the management of some of the most impactful cholangiopathies associated with liver transplantation, such as biliary atresia or ischemic cholangiopathy. Recent advances in tissue engineering and in vitro cholangiocyte culture have made the achievement of this goal possible. Here we provide an overview of these developments and review the progress towards the generation and transplantation of bioengineered bile ducts. This article is part of a Special Issue entitled: Cholangiocytes in Health and Diseaseedited by Jesus Banales, Marco Marzioni and Peter Jansen.
- Published
- 2018
40. Variability of human pluripotent stem cell lines
- Author
-
Daniel Ortmann and Ludovic Vallier
- Subjects
0301 basic medicine ,Cell type ,business.industry ,Somatic cell ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Cell Differentiation ,Biology ,Cellular Reprogramming ,Embryonic stem cell ,Cell Line ,Cell biology ,Biotechnology ,03 medical and health sciences ,030104 developmental biology ,Genetics ,Humans ,Progenitor cell ,business ,Induced pluripotent stem cell ,Cell potency ,Reprogramming ,Embryonic Stem Cells ,Developmental Biology - Abstract
Human pluripotent stem cells derived from embryos (human Embryonic Stem Cells or hESCs) or generated by direct reprogramming of somatic cells (human Induced Pluripotent Stem Cells or hiPSCs) can proliferate almost indefinitely in vitro while maintaining the capacity to differentiate into a broad diversity of cell types. These two properties (self-renewal and pluripotency) confers human pluripotent stem cells a unique interest for clinical applications since they could allow the production of infinite quantities of cells for disease modelling, drug screening and cell based therapy. However, recent studies have clearly established that human pluripotent stem cell lines can display variable capacity to differentiate into specific lineages. Consequently, the development of universal protocols of differentiation which could work efficiently with any human pluripotent cell line is complicated substantially. As a consequence, each protocol needs to be adapted to every cell line thereby limiting large scale applications and precluding personalised therapies. Here, we summarise our knowledge concerning the origin of this variability and describe potential solutions currently available to bypass this major challenge.
- Published
- 2017
41. Genome editing reveals a role for OCT4 in human embryogenesis
- Author
-
Benjamin E. Powell, Jens Kleinjung, Rebecca A. Lea, James M. A. Turner, Alessandro Bertero, Kay Elder, Paul Blakeley, Ludovic Vallier, Kathy K. Niakan, Dagan Wells, Kirsten E. Snijders, N Kubikova, Daesik Kim, Valdone Maciulyte, Afshan McCarthy, Norah M. E. Fogarty, Sissy E. Wamaitha, Jin-Soo Kim, Vallier, Ludovic [0000-0002-3848-2602], Niakan, Kathy [0000-0003-1646-4734], and Apollo - University of Cambridge Repository
- Subjects
Male ,0301 basic medicine ,Homeobox protein NANOG ,Zygote ,Human Embryonic Stem Cells ,Embryonic Development ,Cell fate determination ,Biology ,Substrate Specificity ,Mice ,03 medical and health sciences ,Genome editing ,Ectoderm ,medicine ,Animals ,Humans ,Cell Lineage ,Blastocyst ,Gene ,reproductive and urinary physiology ,Gene Editing ,Genetics ,Regulation of gene expression ,Multidisciplinary ,Gene Expression Regulation, Developmental ,Nanog Homeobox Protein ,Embryo, Mammalian ,Embryonic stem cell ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,embryonic structures ,Female ,CRISPR-Cas Systems ,Octamer Transcription Factor-3 ,Germ Layers - Abstract
Despite their fundamental biological and clinical importance, the molecular mechanisms that regulate the first cell fate decisions in the human embryo are not well understood. Here we use CRISPR-Cas9-mediated genome editing to investigate the function of the pluripotency transcription factor OCT4 during human embryogenesis. We identified an efficient OCT4-targeting guide RNA using an inducible human embryonic stem cell-based system and microinjection of mouse zygotes. Using these refined methods, we efficiently and specifically targeted the gene encoding OCT4 (POU5F1) in diploid human zygotes and found that blastocyst development was compromised. Transcriptomics analysis revealed that, in POU5F1-null cells, gene expression was downregulated not only for extra-embryonic trophectoderm genes, such as CDX2, but also for regulators of the pluripotent epiblast, including NANOG. By contrast, Pou5f1-null mouse embryos maintained the expression of orthologous genes, and blastocyst development was established, but maintenance was compromised. We conclude that CRISPR-Cas9-mediated genome editing is a powerful method for investigating gene function in the context of human development.
- Published
- 2017
42. Genetic association analysis identifies variants associated with disease progression in primary sclerosing cholangitis
- Author
-
Martina Sterneck, Wolfgang Lieb, Christian Rust, Tom H. Karlsen, Massimo Pinzani, Erik Schrumpf, John E. Eaton, Gideon M. Hirschfield, Christoph Schramm, Richard Sandford, Ulrich Beuers, Andrew Mason, Espen Melum, Daniel Gotthardt, A. Boudewijn de Vries, I. Franceschet, Konstantinos N. Lazaridis, Floris Imhann, Marco Carbone, Pietro Invernizzi, Mark S. Silverberg, Simon Hohenester, Maria Consiglia Bragazzi, Andreas Teufel, Bart van Hoek, Martti Färkkilä, Einar Bjornsson, Cyriel Y. Ponsioen, Brijesh Srivastava, Joanne Verheij, Roger W. Chapman, Krista Rombouts, Niklas K. Björkström, Johannes R. Hov, Weiwei Wang, F. Sampaziotis, Ludovic Vallier, Albert Parés, Eleonora A. M. Festen, Kristian Holm, Kalliopi Zachou, Katrin Böttcher, Christopher L. Bowlus, Xiaojun Jiang, Trine Folseraas, Elisabeth M.G. de Vries, Simon M. Rushbrook, Piotr Milkiewicz, Carl A. Anderson, Georgios N. Dalekos, David Ellinghaus, Hanns-Ulrich Marschall, Rinse K. Weersma, Marco Marzioni, Olivier Chazouillères, Domenico Alvaro, Christian Rupp, Angela M. Cheung, Jimmy Z. Liu, Brian D. Juran, Michael P. Manns, Rudi Alberts, Bertus Eksteen, Tobias J. Weismüller, Graeme J.M. Alexander, Annarosa Floreani, Tobias Müller, Stefan Schreiber, Andre Franke, Elizabeth C. Goode, Henrike Lenzen, Arnau Vich Vila, Annika Bergquist, Kirsten Muri Boberg, Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI), Gastroenterology and Hepatology, AGEM - Amsterdam Gastroenterology Endocrinology Metabolism, Graduate School, Pathology, Alberts, R, De Vries, E, Goode, E, Jiang, X, Sampaziotis, F, Rombouts, K, Böttcher, K, Folseraas, T, Weismüller, T, Mason, A, Wang, W, Alexander, G, Alvaro, D, Bergquist, A, Björkström, N, Beuers, U, Björnsson, E, Boberg, K, Bowlus, C, Bragazzi, M, Carbone, M, Chazouillères, O, Cheung, A, Dalekos, G, Eaton, J, Eksteen, B, Ellinghaus, D, Färkkilä, M, Festen, E, Floreani, A, Franceschet, I, Gotthardt, D, Hirschfield, G, Hoek, B, Holm, K, Hohenester, S, Hov, J, Imhann, F, Invernizzi, P, Juran, B, Lenzen, H, Lieb, W, Liu, J, Marschall, H, Marzioni, M, Melum, E, Milkiewicz, P, Müller, T, Pares, A, Rupp, C, Rust, C, Sandford, R, Schramm, C, Schreiber, S, Schrumpf, E, Silverberg, M, Srivastava, B, Sterneck, M, Teufel, A, Vallier, L, Verheij, J, Vila, A, Vries, B, Zachou, K, Chapman, R, Manns, M, Pinzani, M, Rushbrook, S, Lazaridis, K, Franke, A, Anderson, C, Karlsen, T, Ponsioen, C, Weersma, R, Centre of Excellence in Complex Disease Genetics, Doctoral Programme in Drug Research, Department of Medicine, Clinicum, Gastroenterologian yksikkö, HUS Abdominal Center, and Universitat de Barcelona
- Subjects
Male ,0301 basic medicine ,Oncology ,Candidate gene ,Cholangitis ,medicine.medical_treatment ,Medizin ,Trasplantament hepàtic ,Genome-wide association study ,Kaplan-Meier Estimate ,LIVER FIBROSIS ,Liver transplantation ,Bioinformatics ,Sclerosing ,Oral and gastrointestinal ,Primary sclerosing cholangitis ,genetics ,liver transplantation ,Cohort Studies ,ACTIVATION ,0302 clinical medicine ,MED/12 - GASTROENTEROLOGIA ,MULTIPLE ,2.1 Biological and endogenous factors ,EPIDEMIOLOGY ,Aetiology ,CIRRHOSIS ,Bilious diseases and biliousness ,Liver Disease ,digestive, oral, and skin physiology ,Gastroenterology ,Single Nucleotide ,Primary sclerosing cholangiti ,Middle Aged ,3. Good health ,ULCERATIVE-COLITIS ,Disease Progression ,Female ,030211 gastroenterology & hepatology ,Adult ,medicine.medical_specialty ,Cholangitis, Sclerosing ,Chronic Liver Disease and Cirrhosis ,Clinical Sciences ,Malalties del tracte biliar ,Single-nucleotide polymorphism ,HEPATIC STELLATE CELLS ,Polymorphism, Single Nucleotide ,International PSC Study Group ,Article ,Paediatrics and Reproductive Medicine ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Internal medicine ,Genetics ,medicine ,Humans ,Polymorphism ,GENOME-WIDE ASSOCIATION ,Allele ,Digestive Diseases - (Gallbladder) ,Survival analysis ,Proportional Hazards Models ,MALIGNANCY ,The UK PSC Consortium ,Transplantation ,Gastroenterology & Hepatology ,business.industry ,Proportional hazards model ,medicine.disease ,RISK LOCI ,Logistic Models ,030104 developmental biology ,3121 General medicine, internal medicine and other clinical medicine ,genetic ,Hepatic transplantation ,Thrombospondins ,Digestive Diseases ,business ,Genètica - Abstract
ObjectivePrimary sclerosing cholangitis (PSC) is a genetically complex, inflammatory bile duct disease of largely unknown aetiology often leading to liver transplantation or death. Little is known about the genetic contribution to the severity and progression of PSC. The aim of this study is to identify genetic variants associated with PSC disease progression and development of complications.DesignWe collected standardised PSC subphenotypes in a large cohort of 3402 patients with PSC. After quality control, we combined 130 422 single nucleotide polymorphisms of all patients—obtained using the Illumina immunochip—with their disease subphenotypes. Using logistic regression and Cox proportional hazards models, we identified genetic variants associated with binary and time-to-event PSC subphenotypes.ResultsWe identified genetic variant rs853974 to be associated with liver transplant-free survival (p=6.07×10–9). Kaplan-Meier survival analysis showed a 50.9% (95% CI 41.5% to 59.5%) transplant-free survival for homozygous AA allele carriers of rs853974 compared with 72.8% (95% CI 69.6% to 75.7%) for GG carriers at 10 years after PSC diagnosis. For the candidate gene in the region, RSPO3, we demonstrated expression in key liver-resident effector cells, such as human and murine cholangiocytes and human hepatic stellate cells.ConclusionWe present a large international PSC cohort, and report genetic loci associated with PSC disease progression. For liver transplant-free survival, we identified a genome-wide significant signal and demonstrated expression of the candidate gene RSPO3 in key liver-resident effector cells. This warrants further assessments of the role of this potential key PSC modifier gene.
- Published
- 2017
43. Directed differentiation of human induced pluripotent stem cells into functional cholangiocyte-like cells
- Author
-
Nicholas R.F. Hannan, Alessandro Bertero, Fotios Sampaziotis, Ludovic Vallier, Miguel Cardoso de Brito, Imbisaat Geti, Sampaziotis, Fotios [0000-0003-0812-7586], Vallier, Ludovic [0000-0002-3848-2602], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,Stem-cell differentiation ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cholangiocyte ,03 medical and health sciences ,Directed differentiation ,Pluripotent stem cells ,medicine ,Humans ,Progenitor cell ,Induced pluripotent stem cell ,Disease model ,Cell Differentiation ,Epithelial Cells ,Cell biology ,Endothelial stem cell ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Cell culture ,Bile Ducts ,Stem cell ,Endoderm - Abstract
The difficulty in isolating and propagating functional primary cholangiocytes is a major limitation in the study of biliary disorders and the testing of novel therapeutic agents. To overcome this problem, we have developed a platform for the differentiation of human pluripotent stem cells (hPSCs) into functional cholangiocyte-like cells (CLCs). We have previously reported that our 26-d protocol closely recapitulates key stages of biliary development, starting with the differentiation of hPSCs into endoderm and subsequently into foregut progenitor (FP) cells, followed by the generation of hepatoblasts (HBs), cholangiocyte progenitors (CPs) expressing early biliary markers and mature CLCs displaying cholangiocyte functionality. Compared with alternative protocols for biliary differentiation of hPSCs, our system does not require coculture with other cell types and relies on chemically defined conditions up to and including the generation of CPs. A complex extracellular matrix is used for the maturation of CLCs; therefore, experience in hPSC culture and 3D organoid systems may be necessary for optimal results. Finally, the capacity of our platform for generating large amounts of disease-specific functional cholangiocytes will have broad applications for cholangiopathies, in disease modeling and for screening of therapeutic compounds.
- Published
- 2017
44. Cholangiocyte organoids can repair bile ducts after transplantation in the human liver
- Author
-
Sanjay Sinha, Victoria L. Mulcahy, William Gelson, Kourosh Saeb-Parsy, Brandon T Wesley, Sarah A. Teichmann, Laure Gambardella, Timothy E. Beach, Sharayne Robinson, Michael P. Murphy, Paul Gibbs, Stephanie Brown, Richard L. Gieseck, Peter C. Humphreys, Corrina Fear, Anna Osnato, Krishnaa T. Mahbubani, Gareth Corbett, Irum Amin, Olivia C. Tysoe, Daniele Muraro, Christopher J.E. Watson, Espen Melum, Daniel Ortmann, Andrew J. Butler, George F. Mells, Teresa Brevini, Keziah Crick, Sara Upponi, Fotios Sampaziotis, Susan E. Davies, Teik Choon See, Giovanni Canu, Stephen J. Sawiak, Jose Garcia-Bernardo, Edmund Godfrey, Ludovic Vallier, N.L. Berntsen, Lisa Swift, Johannes Bargehr, Sampaziotis, Fotios [0000-0003-0812-7586], Muraro, Daniele [0000-0001-9601-237X], Tysoe, Olivia C [0000-0002-1061-1484], Sawiak, Stephen [0000-0003-4210-9816], Brevini, Teresa [0000-0002-3581-5379], Wesley, Brandon T [0000-0003-0530-329X], Garcia-Bernardo, Jose [0000-0003-3626-5433], Mahbubani, Krishnaa [0000-0002-1327-2334], Canu, Giovanni [0000-0002-3349-4479], Berntsen, Natalie L [0000-0002-5949-8910], Mulcahy, Victoria L [0000-0001-7091-4789], Robinson, Sharayne [0000-0001-6819-3433], Swift, Lisa [0000-0001-9044-688X], Gambardella, Laure [0000-0001-5771-1565], Bargehr, Johannes [0000-0002-9304-3573], Osnato, Anna [0000-0001-5241-1512], Murphy, Michael P [0000-0003-1115-9618], Gibbs, Paul [0000-0003-2193-5377], Sinha, Sanjay [0000-0001-5900-1209], Teichmann, Sarah A [0000-0002-6294-6366], Melum, Espen [0000-0001-6903-6878], Saeb-Parsy, Kourosh [0000-0002-0633-3696], Vallier, Ludovic [0000-0002-3848-2602], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Tissue and Organ Procurement ,medicine.medical_treatment ,Cell- and Tissue-Based Therapy ,Context (language use) ,Bile Duct Diseases ,Biology ,Liver transplantation ,Mesenchymal Stem Cell Transplantation ,Regenerative medicine ,Cholangiocyte ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Organoid ,medicine ,Animals ,Bile ,Humans ,RNA-Seq ,Common Bile Duct ,Multidisciplinary ,Gallbladder ,Epithelial Cells ,Cell biology ,Liver Transplantation ,Transplantation ,Organoids ,030104 developmental biology ,Bile Ducts, Intrahepatic ,Gene Expression Regulation ,Liver ,030211 gastroenterology & hepatology ,Bile Ducts ,Transcriptome ,Ex vivo - Abstract
Organoids regenerate human bile ducts Bile ducts carry bile from the liver and gall bladder to the small intestine, where it aids digestion. Cholangiocytes are epithelial cells that line bile ducts and modify bile as its transported through the biliary tree. Chronic liver diseases involving cholangiocytes account for a large fraction of liver failure and the need for liver transplantation. Because liver donors are in short supply, Sampaziotis et al. used organoid technology to develop a cell-based therapy using human tissue (see the Perspective by Kurial and Willenbring). Cholangiocyte organoids were transplanted into the intrahepatic ducts of deceased human donor livers undergoing ex vivo normothermic perfusion. The livers could be maintained for up to 100 hours, and the transplanted organoids engrafted, exhibited function, and could repair bile ducts. Science , this issue p. 839 ; see also p. 786
- Published
- 2019
45. GDF15 and the beneficial actions of metformin in pre-diabetes
- Author
-
Audrey Melvin, Bernard B. Allan, David B. Savage, David Ron, Debra Rimmington, Giles S.H. Yeo, Sergio Rodriguez-Cuenca, Michael Chen, Frank Reimann, Naveed Sattar, David Preiss, Pranali Taskar, Heather P. Harding, Satish Patel, Emily L. Miedzybrodzka, Antonio Vidal-Puig, Fiona M. Gribble, Ming Yang, Irene Cimino, Deborah A. Goldspink, John Tadross, Anthony P. Coll, Ludovic Vallier, Sam Virtue, Paul Welsh, Y. C. Loraine Tung, Stephen O'Rahilly, and Rute A. Tomaz
- Subjects
medicine.medical_specialty ,Receptor complex ,endocrine system diseases ,medicine.medical_treatment ,030209 endocrinology & metabolism ,Type 2 diabetes ,03 medical and health sciences ,0302 clinical medicine ,Weight loss ,Internal medicine ,Diabetes mellitus ,medicine ,030304 developmental biology ,0303 health sciences ,business.industry ,Insulin ,nutritional and metabolic diseases ,medicine.disease ,3. Good health ,Metformin ,Endocrinology ,GDF15 ,medicine.symptom ,business ,Weight gain ,medicine.drug - Abstract
SummaryMetformin, the world’s most prescribed anti-diabetic drug, is also effective in preventing Type 2 diabetes in people at high risk, by lowering body weight, fat mass and circulating insulin levels through mechanisms that are incompletely understood. Recent observational studies reporting the association of metformin use and circulating levels of GDF15 led us to hypothesize that GDF15, which signals through a specific receptor complex in the hindbrain to reduce body weight, might mediate these effects. We measured GDF15 in people without diabetes from a randomized placebo-controlled trial of metformin. Over 18 months, participants allocated metformin lost significant weight and levels of GDF15 were persistently elevated compared to placebo. The change in plasma GDF15 in this study correlated positively with weight loss. In wild-type mice, oral metformin increased circulating GDF15 with GDF15 expression increasing predominantly in the distal intestine and the kidney. Metformin prevented weight gain in response to high fat diet in wild-type mice but not in mice lacking GDF15 or its receptor GFRAL. In obese, high fat-fed mice, the effects of metformin to reduce body weight were reversed by a GFRAL antagonist antibody. Metformin had effects on both energy intake and energy expenditure that required GDF15. The insulin sensitising effects of metformin determined by insulin tolerance were abolished in mice lacking GDF15. Metformin significantly reduced fasting glucose and insulin levels in wild type but not in mice lacking GDF15. In summary, metformin increases the circulating levels of GDF15, which appears to be necessary for many of its actions as a metabolic chemopreventive agent.
- Published
- 2019
46. Tissue-Engineering the Intestine: The Trials before the Trials
- Author
-
Matthias P. Lutolf, Vivian S. W. Li, Kim B. Jensen, Ryan K. Conder, Paolo De Coppi, Sarah Chan, Tracy C. Grikscheit, Ludovic Vallier, Hans Clevers, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
Short Bowel Syndrome ,Scaffold ,media_common.quotation_subject ,Organogenesis ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,Design strategy ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Tissue scaffolds ,Tissue engineering ,Genetics ,medicine ,Animals ,Humans ,Function (engineering) ,030304 developmental biology ,media_common ,Cell Proliferation ,0303 health sciences ,Tissue Engineering ,Tissue Scaffolds ,Cell Differentiation ,Cell Biology ,Short bowel syndrome ,medicine.disease ,Intestines ,Organoids ,Risk analysis (engineering) ,Molecular Medicine ,030217 neurology & neurosurgery - Abstract
Building complex tissues requires the development of innovative interdisciplinary engineering solutions. In this Forum, the INTENS Consortium discuss experimental considerations and challenges for generating a tissue-engineered intestine for the treatment of short bowel syndrome, taking into account cell source, scaffold choice, and design strategy for achieving proper assembly and function.
- Published
- 2019
47. Isolation and propagation of primary human cholangiocyte organoids for the generation of bioengineered biliary tissue
- Author
-
Ludovic Vallier, Krishnaa T. Mahbubani, Espen Melum, Alexander W. Justin, Si Emma Chen, Olivia C. Tysoe, Hajer Zedira, Teresa Brevini, Athina E. Markaki, Anna Katharina Frank, Kourosh Saeb-Parsy, Fotios Sampaziotis, Frank, Anna K [0000-0003-1378-5129], Sampaziotis, Fotios [0000-0003-0812-7586], and Apollo - University of Cambridge Repository
- Subjects
medicine.medical_treatment ,Biocompatible Materials ,Cell Separation ,Biology ,Liver transplantation ,Regenerative medicine ,General Biochemistry, Genetics and Molecular Biology ,Cholangiocyte ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Tissue engineering ,Organoid ,medicine ,Animals ,Humans ,Regeneration ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Tissue Engineering ,Tissue Scaffolds ,Regeneration (biology) ,Equipment Design ,3. Good health ,Cell biology ,Transplantation ,Organoids ,Bile Ducts ,030217 neurology & neurosurgery ,Adult stem cell - Abstract
Pediatric liver transplantation is often required as a consequence of biliary disorders because of the lack of alternative treatments for repairing or replacing damaged bile ducts. To address the lack of availability of pediatric livers suitable for transplantation, we developed a protocol for generating bioengineered biliary tissue suitable for biliary reconstruction. Our platform allows the derivation of cholangiocyte organoids (COs) expressing key biliary markers and retaining functions of primary extra- or intrahepatic duct cholangiocytes within 2 weeks of isolation. COs are subsequently seeded on polyglycolic acid (PGA) scaffolds or densified collagen constructs for 4 weeks to generate bioengineered tissue retaining biliary characteristics. Expertise in organoid culture and tissue engineering is desirable for optimal results. COs correspond to mature functional cholangiocytes, differentiating our method from alternative organoid systems currently available that propagate adult stem cells. Consequently, COs provide a unique platform for studies in biliary physiology and pathophysiology, and the resulting bioengineered tissue has broad applications for regenerative medicine and cholangiopathies.
- Published
- 2019
48. Use of Biliary Organoids in Cholestasis Research
- Author
-
Fotios, Sampaziotis, Olivia, Tysoe, Teresa, Brevini, and Ludovic, Vallier
- Subjects
Organoids ,Cholestasis ,Liver ,Induced Pluripotent Stem Cells ,Gallbladder ,Humans ,Bile Ducts - Abstract
Cholangiocytes play a crucial role in the pathophysiology of cholestasis. However, research on human cholangiocytes has been restricted by challenges in long-term propagation and large-scale expansion of primary biliary epithelium. The advent of organoid technology has overcome this limitation allowing long-term culture of a variety of epithelia from multiple organs. Here, we describe two methods for growing human cholangiocytes in organoid format. The first applies to the generation of intrahepatic bile ducts using human induced pluripotent stem cells using a protocol of differentiation that recapitulates physiological bile duct development. The second method allows the propagation of primary biliary epithelium from the extrahepatic ducts or gallbladder. Both protocols result in large numbers of cholangiocyte organoids expressing biliary markers and maintaining key cholangiocyte functions.
- Published
- 2019
49. Conditional Gene Knockout in Human Cells with Inducible CRISPR/Cas9
- Author
-
Kirsten E, Snijders, James D, Cooper, Ludovic, Vallier, and Alessandro, Bertero
- Subjects
Pluripotent Stem Cells ,Gene Knockout Techniques ,Humans ,CRISPR-Cas Systems ,RNA, Guide, Kinetoplastida - Abstract
The advent of the easily programmable and efficient CRISPR/Cas9 nuclease system has revolutionized genetic engineering. While conventional gene knockout experiments using CRISPR/Cas9 are very valuable, these are not well suited to study stage-specific gene function in dynamic situations such as development or disease. Here we describe a CRISPR/Cas9-based OPTimized inducible gene KnockOut method (OPTiKO) for conditional loss-of-function studies in human cells. This approach relies on an improved tetracycline-inducible system for conditional expression of single guide RNAs (sgRNAs) that drive Cas9 activity. In order to ensure homogeneous and stable expression, the necessary transgenes are expressed following rapid and efficient single-step genetic engineering of the AAVS1 genomic safe harbor. When implemented in human pluripotent stem cells (hPSCs), the approach can be then efficiently applied to virtually any hPSC-derived human cell type at various stages of development or disease.
- Published
- 2019
50. A proteomic time course through the differentiation of human induced pluripotent stem cells into hepatocyte-like cells
- Author
-
Charis-Patricia Segeritz, Ludovic Vallier, Kathryn S. Lilley, A.D. Cromarty, Tracey Hurrell, Cromarty, Allan D [0000-0002-9512-6081], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Proteomics ,Quantitative proteomics ,Induced Pluripotent Stem Cells ,lcsh:Medicine ,Biology ,Regenerative medicine ,Article ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,lcsh:Science ,Multidisciplinary ,lcsh:R ,Cell Differentiation ,Phenotype ,Antigens, Differentiation ,In vitro ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Hepatocyte ,Proteome ,Hepatocytes ,lcsh:Q ,030217 neurology & neurosurgery - Abstract
Numerous in vitro models endeavour to mimic the characteristics of primary human hepatocytes for applications in regenerative medicine and pharmaceutical science. Mature hepatocyte-like cells (HLCs) derived from human induced pluripotent stem cells (hiPSCs) are one such in vitro model. Due to insufficiencies in transcriptome to proteome correlation, characterising the proteome of HLCs is essential to provide a suitable framework for their continual optimization. Here we interrogated the proteome during stepwise differentiation of hiPSCs into HLCs over 40 days. Whole cell protein lysates were collected and analysed using stabled isotope labelled mass spectrometry based proteomics. Quantitative proteomics identified over 6,000 proteins in duplicate multiplexed labelling experiments across two different time course series. Inductive cues in differentiation promoted sequential acquisition of hepatocyte specific markers. Analysis of proteins classically assigned as hepatic markers demonstrated trends towards maximum relative abundance between differentiation day 30 and 32. Characterisation of abundant proteins in whole cells provided evidence of the time dependent transition towards proteins corresponding with the functional repertoire of the liver. This data highlights how far the proteome of undifferentiated precursors have progressed to acquire a hepatic phenotype and constructs a platform for optimisation and improved maturation of HLC differentiation.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.