51
|
Villa CE, Cheroni C, Dotter CP, López-Tóbon A, Oliveira B, Sacco R, Yahya AÇ, Morandell J, Gabriele M, Tavakoli MR, Lyudchik J, Sommer C, Gabitto M, Danzl JG, Testa G, Novarino G. CHD8 haploinsufficiency links autism to transient alterations in excitatory and inhibitory trajectories. Cell Rep 2022; 39:110615. [PMID: 35385734 DOI: 10.1016/j.celrep.2022.110615] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 11/18/2021] [Accepted: 03/13/2022] [Indexed: 12/13/2022] Open
Abstract
Mutations in the chromodomain helicase DNA-binding 8 (CHD8) gene are a frequent cause of autism spectrum disorder (ASD). While its phenotypic spectrum often encompasses macrocephaly, implicating cortical abnormalities, how CHD8 haploinsufficiency affects neurodevelopmental is unclear. Here, employing human cerebral organoids, we find that CHD8 haploinsufficiency disrupted neurodevelopmental trajectories with an accelerated and delayed generation of, respectively, inhibitory and excitatory neurons that yields, at days 60 and 120, symmetrically opposite expansions in their proportions. This imbalance is consistent with an enlargement of cerebral organoids as an in vitro correlate of patients' macrocephaly. Through an isogenic design of patient-specific mutations and mosaic organoids, we define genotype-phenotype relationships and uncover their cell-autonomous nature. Our results define cell-type-specific CHD8-dependent molecular defects related to an abnormal program of proliferation and alternative splicing. By identifying cell-type-specific effects of CHD8 mutations, our study uncovers reproducible developmental alterations that may be employed for neurodevelopmental disease modeling.
Collapse
Affiliation(s)
- Carlo Emanuele Villa
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy
| | - Cristina Cheroni
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
| | - Christoph P Dotter
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Alejandro López-Tóbon
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
| | - Bárbara Oliveira
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Roberto Sacco
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Aysan Çerağ Yahya
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Jasmin Morandell
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Michele Gabriele
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy
| | - Mojtaba R Tavakoli
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Julia Lyudchik
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Christoph Sommer
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | | | - Johann G Danzl
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Giuseppe Testa
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy.
| | - Gaia Novarino
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria.
| |
Collapse
|
52
|
Ray-Gallet D, Almouzni G. H3–H4 histone chaperones and cancer. Curr Opin Genet Dev 2022; 73:101900. [DOI: 10.1016/j.gde.2022.101900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 12/16/2022]
|
53
|
Sampson DM, Dubis AM, Chen FK, Zawadzki RJ, Sampson DD. Towards standardizing retinal optical coherence tomography angiography: a review. LIGHT, SCIENCE & APPLICATIONS 2022; 11:63. [PMID: 35304441 PMCID: PMC8933532 DOI: 10.1038/s41377-022-00740-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 02/01/2022] [Accepted: 02/14/2022] [Indexed: 05/11/2023]
Abstract
The visualization and assessment of retinal microvasculature are important in the study, diagnosis, monitoring, and guidance of treatment of ocular and systemic diseases. With the introduction of optical coherence tomography angiography (OCTA), it has become possible to visualize the retinal microvasculature volumetrically and without a contrast agent. Many lab-based and commercial clinical instruments, imaging protocols and data analysis methods and metrics, have been applied, often inconsistently, resulting in a confusing picture that represents a major barrier to progress in applying OCTA to reduce the burden of disease. Open data and software sharing, and cross-comparison and pooling of data from different studies are rare. These inabilities have impeded building the large databases of annotated OCTA images of healthy and diseased retinas that are necessary to study and define characteristics of specific conditions. This paper addresses the steps needed to standardize OCTA imaging of the human retina to address these limitations. Through review of the OCTA literature, we identify issues and inconsistencies and propose minimum standards for imaging protocols, data analysis methods, metrics, reporting of findings, and clinical practice and, where this is not possible, we identify areas that require further investigation. We hope that this paper will encourage the unification of imaging protocols in OCTA, promote transparency in the process of data collection, analysis, and reporting, and facilitate increasing the impact of OCTA on retinal healthcare delivery and life science investigations.
Collapse
Affiliation(s)
- Danuta M Sampson
- Surrey Biophotonics, Centre for Vision, Speech and Signal Processing and School of Biosciences and Medicine, The University of Surrey, Guildford, GU2 7XH, UK.
| | - Adam M Dubis
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Trust and UCL Institute of Ophthalmology, London, EC1V 2PD, UK
| | - Fred K Chen
- Centre for Ophthalmology and Visual Science (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, Western Australia, 6009, Australia
- Department of Ophthalmology, Royal Perth Hospital, Perth, Western Australia, 6000, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, 3002, Australia
| | - Robert J Zawadzki
- Department of Ophthalmology & Vision Science, University of California Davis, Sacramento, CA, 95817, USA
| | - David D Sampson
- Surrey Biophotonics, Advanced Technology Institute, School of Physics and School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| |
Collapse
|
54
|
Gaglia G, Kabraji S, Rammos D, Dai Y, Verma A, Wang S, Mills CE, Chung M, Bergholz JS, Coy S, Lin JR, Jeselsohn R, Metzger O, Winer EP, Dillon DA, Zhao JJ, Sorger PK, Santagata S. Temporal and spatial topography of cell proliferation in cancer. Nat Cell Biol 2022; 24:316-326. [PMID: 35292783 PMCID: PMC8959396 DOI: 10.1038/s41556-022-00860-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 01/31/2022] [Indexed: 02/06/2023]
Abstract
Proliferation is a fundamental trait of cancer cells, but its properties and spatial organization in tumours are poorly characterized. Here we use highly multiplexed tissue imaging to perform single-cell quantification of cell cycle regulators and then develop robust, multivariate, proliferation metrics. Across diverse cancers, proliferative architecture is organized at two spatial scales: large domains, and smaller niches enriched for specific immune lineages. Some tumour cells express cell cycle regulators in the (canonical) patterns expected of freely growing cells, a phenomenon we refer to as 'cell cycle coherence'. By contrast, the cell cycles of other tumour cell populations are skewed towards specific phases or exhibit non-canonical (incoherent) marker combinations. Coherence varies across space, with changes in oncogene activity and therapeutic intervention, and is associated with aggressive tumour behaviour. Thus, multivariate measures from high-plex tissue images capture clinically significant features of cancer proliferation, a fundamental step in enabling more precise use of anti-cancer therapies.
Collapse
Affiliation(s)
- Giorgio Gaglia
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sheheryar Kabraji
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA.
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA.
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Danae Rammos
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yang Dai
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ana Verma
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shu Wang
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA
| | - Caitlin E Mills
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Mirra Chung
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Johann S Bergholz
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Shannon Coy
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jia-Ren Lin
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | - Rinath Jeselsohn
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Otto Metzger
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Eric P Winer
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Deborah A Dillon
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jean J Zhao
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | - Sandro Santagata
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Dana Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
55
|
Schapiro D, Yapp C, Sokolov A, Reynolds SM, Chen YA, Sudar D, Xie Y, Muhlich J, Arias-Camison R, Arena S, Taylor AJ, Nikolov M, Tyler M, Lin JR, Burlingame EA, Chang YH, Farhi SL, Thorsson V, Venkatamohan N, Drewes JL, Pe'er D, Gutman DA, Herrmann MD, Gehlenborg N, Bankhead P, Roland JT, Herndon JM, Snyder MP, Angelo M, Nolan G, Swedlow JR, Schultz N, Merrick DT, Mazzili SA, Cerami E, Rodig SJ, Santagata S, Sorger PK. MITI minimum information guidelines for highly multiplexed tissue images. Nat Methods 2022; 19:262-267. [PMID: 35277708 PMCID: PMC9009186 DOI: 10.1038/s41592-022-01415-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The imminent release of tissue atlases combining multi-channel microscopy with single cell sequencing and other omics data from normal and diseased specimens creates an urgent need for data and metadata standards that guide data deposition, curation and release. We describe a Minimum Information about highly multiplexed Tissue Imaging (MITI) standard that applies best practices developed for genomics and other microscopy data to highly multiplexed tissue images and traditional histology.
Collapse
Affiliation(s)
- Denis Schapiro
- Laboratory of Systems Pharmacology, Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University Hospital and Heidelberg University, Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Clarence Yapp
- Laboratory of Systems Pharmacology, Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
- Image and Data Analysis Core, Harvard Medical School, Boston, MA, USA
| | - Artem Sokolov
- Laboratory of Systems Pharmacology, Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | | | - Yu-An Chen
- Laboratory of Systems Pharmacology, Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
| | - Damir Sudar
- Quantitative Imaging Systems LLC, Portland, OR, USA
| | - Yubin Xie
- Program in Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeremy Muhlich
- Laboratory of Systems Pharmacology, Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
| | - Raquel Arias-Camison
- Laboratory of Systems Pharmacology, Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
| | - Sarah Arena
- Laboratory of Systems Pharmacology, Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
| | | | | | - Madison Tyler
- Laboratory of Systems Pharmacology, Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
| | - Jia-Ren Lin
- Laboratory of Systems Pharmacology, Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
| | - Erik A Burlingame
- Oregon Health and Science University, Portland, OR, USA
- Indica Labs, Albuquerque, NM, USA
| | - Young H Chang
- Oregon Health and Science University, Portland, OR, USA
| | - Samouil L Farhi
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Julia L Drewes
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dana Pe'er
- Program in Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Markus D Herrmann
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Nils Gehlenborg
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Peter Bankhead
- Edinburgh Pathology, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Joseph T Roland
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - John M Herndon
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Michael Angelo
- School of Medicine, Stanford University, Stanford, CA, USA
| | - Garry Nolan
- School of Medicine, Stanford University, Stanford, CA, USA
| | - Jason R Swedlow
- Division of Computational Biology and Centre for Gene Regulation and Expression, University of Dundee, Dundee, UK
| | - Nikolaus Schultz
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Sandro Santagata
- Laboratory of Systems Pharmacology, Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
56
|
Caporale N, Leemans M, Birgersson L, Germain PL, Cheroni C, Borbély G, Engdahl E, Lindh C, Bressan RB, Cavallo F, Chorev NE, D'Agostino GA, Pollard SM, Rigoli MT, Tenderini E, Tobon AL, Trattaro S, Troglio F, Zanella M, Bergman Å, Damdimopoulou P, Jönsson M, Kiess W, Kitraki E, Kiviranta H, Nånberg E, Öberg M, Rantakokko P, Rudén C, Söder O, Bornehag CG, Demeneix B, Fini JB, Gennings C, Rüegg J, Sturve J, Testa G. From cohorts to molecules: Adverse impacts of endocrine disrupting mixtures. Science 2022; 375:eabe8244. [PMID: 35175820 DOI: 10.1126/science.abe8244] [Citation(s) in RCA: 124] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Convergent evidence associates exposure to endocrine disrupting chemicals (EDCs) with major human diseases, even at regulation-compliant concentrations. This might be because humans are exposed to EDC mixtures, whereas chemical regulation is based on a risk assessment of individual compounds. Here, we developed a mixture-centered risk assessment strategy that integrates epidemiological and experimental evidence. We identified that exposure to an EDC mixture in early pregnancy is associated with language delay in offspring. At human-relevant concentrations, this mixture disrupted hormone-regulated and disease-relevant regulatory networks in human brain organoids and in the model organisms Xenopus leavis and Danio rerio, as well as behavioral responses. Reinterrogating epidemiological data, we found that up to 54% of the children had prenatal exposures above experimentally derived levels of concern, reaching, for the upper decile compared with the lowest decile of exposure, a 3.3 times higher risk of language delay.
Collapse
Affiliation(s)
- Nicolò Caporale
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy.,Human Technopole, V.le Rita Levi-Montalcini, 1, 20157 Milan, Italy
| | - Michelle Leemans
- UMR 7221, Phyma, CNRS-Muséum National d'Histoire Naturelle, Sorbonne Université, 75005 Paris, France
| | - Lina Birgersson
- Department of Biological and Environmental Sciences, University of Gothenburg, 41463 Gothenburg, Sweden
| | - Pierre-Luc Germain
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Cristina Cheroni
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy.,Human Technopole, V.le Rita Levi-Montalcini, 1, 20157 Milan, Italy
| | - Gábor Borbély
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden
| | - Elin Engdahl
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden.,Department of Organismal Biology, Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Christian Lindh
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, SE-221 85 Lund, Sweden
| | - Raul Bardini Bressan
- Medical Research Council Centre for Regenerative Medicine and Edinburgh Cancer Research UK Centre, University of Edinburgh, Edinburgh, UK
| | - Francesca Cavallo
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Nadav Even Chorev
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Giuseppe Alessandro D'Agostino
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Steven M Pollard
- Medical Research Council Centre for Regenerative Medicine and Edinburgh Cancer Research UK Centre, University of Edinburgh, Edinburgh, UK
| | - Marco Tullio Rigoli
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
| | - Erika Tenderini
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Alejandro Lopez Tobon
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Sebastiano Trattaro
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
| | - Flavia Troglio
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Matteo Zanella
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Åke Bergman
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden.,Department of Environmental Science, Stockholm University, SE-10691 Stockholm, Sweden.,School of Science and Technology, Örebro University, SE-70182 Örebro, Sweden
| | - Pauliina Damdimopoulou
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden.,Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Maria Jönsson
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Wieland Kiess
- Hospital for Children and Adolescents, Department of Women and Child Health, University Hospital, University of Leipzig, 04103 Leipzig, Germany
| | - Efthymia Kitraki
- Lab of Basic Sciences, Faculty of Dentistry, National and Kapodistrian University of Athens, 152 72 Athens, Greece
| | - Hannu Kiviranta
- Department of Health Security, Finnish Institute for Health and Welfare (THL), Kuopio 70210, Finland
| | - Eewa Nånberg
- School of Health Sciences, Örebro University, SE-70182 Örebro, Sweden
| | - Mattias Öberg
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden.,Institute of Environmental Medicine, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Panu Rantakokko
- Department of Health Security, Finnish Institute for Health and Welfare (THL), Kuopio 70210, Finland
| | - Christina Rudén
- Department of Environmental Science, Stockholm University, SE-10691 Stockholm, Sweden
| | - Olle Söder
- Department of Women's and Children's Health, Pediatric Endocrinology Division, Karolinska Institutet and University Hospital, SE-17176 Stockholm, Sweden
| | - Carl-Gustaf Bornehag
- Faculty of Health, Science and Technology, Department of Health Sciences, Karlstad University, SE- 651 88 Karlstad, Sweden.,Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Barbara Demeneix
- UMR 7221, Phyma, CNRS-Muséum National d'Histoire Naturelle, Sorbonne Université, 75005 Paris, France
| | - Jean-Baptiste Fini
- UMR 7221, Phyma, CNRS-Muséum National d'Histoire Naturelle, Sorbonne Université, 75005 Paris, France
| | - Chris Gennings
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joëlle Rüegg
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden.,Department of Organismal Biology, Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Joachim Sturve
- Department of Biological and Environmental Sciences, University of Gothenburg, 41463 Gothenburg, Sweden
| | - Giuseppe Testa
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy.,Human Technopole, V.le Rita Levi-Montalcini, 1, 20157 Milan, Italy
| |
Collapse
|
57
|
Huizing GJ, Peyré G, Cantini L. Optimal transport improves cell-cell similarity inference in single-cell omics data. Bioinformatics 2022; 38:2169-2177. [PMID: 35157031 PMCID: PMC9004651 DOI: 10.1093/bioinformatics/btac084] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/17/2021] [Accepted: 02/08/2022] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION High-throughput single-cell molecular profiling is revolutionizing biology and medicine by unveiling the diversity of cell types and states contributing to development and disease. The identification and characterization of cellular heterogeneity are typically achieved through unsupervised clustering, which crucially relies on a similarity metric. RESULTS We here propose the use of Optimal Transport (OT) as a cell-cell similarity metric for single-cell omics data. OT defines distances to compare high-dimensional data represented as probability distributions. To speed up computations and cope with the high dimensionality of single-cell data, we consider the entropic regularization of the classical OT distance. We then extensively benchmark OT against state-of-the-art metrics over 13 independent datasets, including simulated, scRNA-seq, scATAC-seq and single-cell DNA methylation data. First, we test the ability of the metrics to detect the similarity between cells belonging to the same groups (e.g. cell types, cell lines of origin). Then, we apply unsupervised clustering and test the quality of the resulting clusters. OT is found to improve cell-cell similarity inference and cell clustering in all simulated and real scRNA-seq data, as well as in scATAC-seq and single-cell DNA methylation data. AVAILABILITY AND IMPLEMENTATION All our analyses are reproducible through the OT-scOmics Jupyter notebook available at https://github.com/ComputationalSystemsBiology/OT-scOmics. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | - Gabriel Peyré
- Département de Mathématiques et Applications de l’Ecole Normale Supérieure, CNRS, Ecole Normale Supérieure, Université PSL, 75005 Paris, France
| | | |
Collapse
|
58
|
Martínez-García M, Hernández-Lemus E. Data Integration Challenges for Machine Learning in Precision Medicine. Front Med (Lausanne) 2022; 8:784455. [PMID: 35145977 PMCID: PMC8821900 DOI: 10.3389/fmed.2021.784455] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
A main goal of Precision Medicine is that of incorporating and integrating the vast corpora on different databases about the molecular and environmental origins of disease, into analytic frameworks, allowing the development of individualized, context-dependent diagnostics, and therapeutic approaches. In this regard, artificial intelligence and machine learning approaches can be used to build analytical models of complex disease aimed at prediction of personalized health conditions and outcomes. Such models must handle the wide heterogeneity of individuals in both their genetic predisposition and their social and environmental determinants. Computational approaches to medicine need to be able to efficiently manage, visualize and integrate, large datasets combining structure, and unstructured formats. This needs to be done while constrained by different levels of confidentiality, ideally doing so within a unified analytical architecture. Efficient data integration and management is key to the successful application of computational intelligence approaches to medicine. A number of challenges arise in the design of successful designs to medical data analytics under currently demanding conditions of performance in personalized medicine, while also subject to time, computational power, and bioethical constraints. Here, we will review some of these constraints and discuss possible avenues to overcome current challenges.
Collapse
Affiliation(s)
- Mireya Martínez-García
- Clinical Research Division, National Institute of Cardiology ‘Ignacio Chávez’, Mexico City, Mexico
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico
- Center for Complexity Sciences, Universidad Nacional Autnoma de Mexico, Mexico City, Mexico
| |
Collapse
|
59
|
Betsholtz C. Toward a granular molecular-anatomic map of the blood vasculature - single-cell RNA sequencing makes the leap. Ups J Med Sci 2022; 127:9051. [PMID: 36337278 PMCID: PMC9602202 DOI: 10.48101/ujms.v127.9051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Single-cell RNA sequencing (scRNAseq) marks the birth of a new era in physiology and medicine. Within foreseeable future, we will know exactly what genes are expressed - and at what levels - in all the different cell types and subtypes that make up our bodies. We will also learn how a particular cell state, whether it occurs during development, tissue repair, or disease, reflects precise changes in gene expression. While profoundly impacting all areas of life science, scRNAseq may lead to a particular leap in vascular biology research. Blood vessels pervade and fulfill essential functions in all organs, but the functions differ. Innumerable organ-specific vascular adaptations and specializations are required. These, in turn, are dictated by differential gene expression by the two principal cellular building blocks of blood vessels: endothelial cells and mural cells. An organotypic vasculature is essential for functions as diverse as thinking, gas exchange, urine excretion, and xenobiotic detoxification in the brain, lung, kidney, and liver, respectively. In addition to the organotypicity, vascular cells also differ along the vascular arterio-venous axis, referred to as zonation, differences that are essential for the regulation of blood pressure and flow. Moreover, gene expression-based molecular changes dictate states of cellular activity, necessary for angiogenesis, vascular permeability, and immune cell trafficking, i.e. functions necessary for development, inflammation, and repair. These different levels of cellular heterogeneity create a nearly infinite phenotypic diversity among vascular cells. In this review, I summarize and exemplify what scRNAseq has brought to the picture in just a few years and point out where it will take us.
Collapse
Affiliation(s)
- Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine-Huddinge, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
60
|
Mohammed Salih M, Carpenter S. What sequencing technologies can teach us about innate immunity. Immunol Rev 2022; 305:9-28. [PMID: 34747035 PMCID: PMC8865538 DOI: 10.1111/imr.13033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/22/2021] [Accepted: 10/16/2021] [Indexed: 01/03/2023]
Abstract
For years, we have taken a reductionist approach to understanding gene regulation through the study of one gene in one cell at a time. While this approach has been fruitful it is laborious and fails to provide a global picture of what is occurring in complex situations involving tightly coordinated immune responses. The emergence of whole-genome techniques provides a system-level view of a response and can provide a plethora of information on events occurring in a cell from gene expression changes to splicing changes and chemical modifications. As with any technology, this often results in more questions than answers, but this wealth of knowledge is providing us with an unprecedented view of what occurs inside our cells during an immune response. In this review, we will discuss the current RNA-sequencing technologies and what they are helping us learn about the innate immune system.
Collapse
Affiliation(s)
- Mays Mohammed Salih
- Department of Molecular Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, USA
| | - Susan Carpenter
- Department of Molecular Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, USA
| |
Collapse
|
61
|
Kasikci Y, Gronemeyer H. Complexity against current cancer research - are we on the wrong track? Int J Cancer 2021; 150:1569-1578. [PMID: 34921726 DOI: 10.1002/ijc.33912] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 11/09/2022]
Abstract
Cancer genetics has led to major discoveries, including proto-oncogene and tumor-suppressor concepts, and cancer genomics generated concepts like driver and passenger genes, revealed tumor heterogeneity and clonal evolution. Reconstructing trajectories of tumorigenesis using spatial and single-cell genomics is possible. Patient stratification and prognostic parameters have been improved. Yet, despite these advances, successful translation into targeted therapies has been scarce and mostly limited to kinase inhibitors. Here, we argue that current cancer research may be on the wrong track, by considering cancer more as a "monogenic" disease, trying to extract common information from thousands of patients, while not properly considering complexity and individual diversity. We suggest to empower a systems cancer approach which reconstructs the information network that has been altered by the tumorigenic events, to analyze hierarchies and predict (druggable) key nodes that could interfere with/block the aberrant information transfer. We also argue that the inter-individual variability between patients of similar cohorts is too high to extract common polygenic network information from large numbers of patients and argue in favor of an individualized approach. The analysis we propose would require a structured multinational and multidisciplinary effort, in which clinicians, and cancer, developmental, cell and computational biologists together with mathematicians and informaticians develop dynamic regulatory networks which integrate the entire information transfer in and between cells and organs in (patho)physiological conditions, revealing hierarchies and available drugs to interfere with key regulators. Based on this blueprint, the altered information transfer in individual cancers could be modeled and possible targeted (combo)therapies proposed. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yasenya Kasikci
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics and Cancer, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Hinrich Gronemeyer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Functional Genomics and Cancer, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France.,Université de Strasbourg, Illkirch, France
| |
Collapse
|
62
|
Li B, Ritchie MD. From GWAS to Gene: Transcriptome-Wide Association Studies and Other Methods to Functionally Understand GWAS Discoveries. Front Genet 2021; 12:713230. [PMID: 34659337 PMCID: PMC8515949 DOI: 10.3389/fgene.2021.713230] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Since their inception, genome-wide association studies (GWAS) have identified more than a hundred thousand single nucleotide polymorphism (SNP) loci that are associated with various complex human diseases or traits. The majority of GWAS discoveries are located in non-coding regions of the human genome and have unknown functions. The valley between non-coding GWAS discoveries and downstream affected genes hinders the investigation of complex disease mechanism and the utilization of human genetics for the improvement of clinical care. Meanwhile, advances in high-throughput sequencing technologies reveal important genomic regulatory roles that non-coding regions play in the transcriptional activities of genes. In this review, we focus on data integrative bioinformatics methods that combine GWAS with functional genomics knowledge to identify genetically regulated genes. We categorize and describe two types of data integrative methods. First, we describe fine-mapping methods. Fine-mapping is an exploratory approach that calibrates likely causal variants underneath GWAS signals. Fine-mapping methods connect GWAS signals to potentially causal genes through statistical methods and/or functional annotations. Second, we discuss gene-prioritization methods. These are hypothesis generating approaches that evaluate whether genetic variants regulate genes via certain genetic regulatory mechanisms to influence complex traits, including colocalization, mendelian randomization, and the transcriptome-wide association study (TWAS). TWAS is a gene-based association approach that investigates associations between genetically regulated gene expression and complex diseases or traits. TWAS has gained popularity over the years due to its ability to reduce multiple testing burden in comparison to other variant-based analytic approaches. Multiple types of TWAS methods have been developed with varied methodological designs and biological hypotheses over the past 5 years. We dive into discussions of how TWAS methods differ in many aspects and the challenges that different TWAS methods face. Overall, TWAS is a powerful tool for identifying complex trait-associated genes. With the advent of single-cell sequencing, chromosome conformation capture, gene editing technologies, and multiplexing reporter assays, we are expecting a more comprehensive understanding of genomic regulation and genetically regulated genes underlying complex human diseases and traits in the future.
Collapse
Affiliation(s)
- Binglan Li
- Department of Biomedical Data Science, Stanford University, Stanford, CA, United States
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States.,Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
63
|
Uhlitz F, Bischoff P, Peidli S, Sieber A, Trinks A, Lüthen M, Obermayer B, Blanc E, Ruchiy Y, Sell T, Mamlouk S, Arsie R, Wei T, Klotz‐Noack K, Schwarz RF, Sawitzki B, Kamphues C, Beule D, Landthaler M, Sers C, Horst D, Blüthgen N, Morkel M. Mitogen-activated protein kinase activity drives cell trajectories in colorectal cancer. EMBO Mol Med 2021; 13:e14123. [PMID: 34409732 PMCID: PMC8495451 DOI: 10.15252/emmm.202114123] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 01/07/2023] Open
Abstract
In colorectal cancer, oncogenic mutations transform a hierarchically organized and homeostatic epithelium into invasive cancer tissue lacking visible organization. We sought to define transcriptional states of colorectal cancer cells and signals controlling their development by performing single-cell transcriptome analysis of tumors and matched non-cancerous tissues of twelve colorectal cancer patients. We defined patient-overarching colorectal cancer cell clusters characterized by differential activities of oncogenic signaling pathways such as mitogen-activated protein kinase and oncogenic traits such as replication stress. RNA metabolic labeling and assessment of RNA velocity in patient-derived organoids revealed developmental trajectories of colorectal cancer cells organized along a mitogen-activated protein kinase activity gradient. This was in contrast to normal colon organoid cells developing along graded Wnt activity. Experimental targeting of EGFR-BRAF-MEK in cancer organoids affected signaling and gene expression contingent on predictive KRAS/BRAF mutations and induced cell plasticity overriding default developmental trajectories. Our results highlight directional cancer cell development as a driver of non-genetic cancer cell heterogeneity and re-routing of trajectories as a response to targeted therapy.
Collapse
Affiliation(s)
- Florian Uhlitz
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- IRI Life SciencesHumboldt University of BerlinBerlinGermany
- German Cancer Consortium (DKTK) Partner Site BerlinGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Philip Bischoff
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Stefan Peidli
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- IRI Life SciencesHumboldt University of BerlinBerlinGermany
| | - Anja Sieber
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- IRI Life SciencesHumboldt University of BerlinBerlinGermany
| | - Alexandra Trinks
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- BIH Bioportal Single CellsBerlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
| | - Mareen Lüthen
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- German Cancer Consortium (DKTK) Partner Site BerlinGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Benedikt Obermayer
- Core Unit Bioinformatics (CUBI)Berlin Institute of Health at Charité Universitätsmedizin – BerlinBerlinGermany
| | - Eric Blanc
- Core Unit Bioinformatics (CUBI)Berlin Institute of Health at Charité Universitätsmedizin – BerlinBerlinGermany
| | - Yana Ruchiy
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Thomas Sell
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- IRI Life SciencesHumboldt University of BerlinBerlinGermany
| | - Soulafa Mamlouk
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- German Cancer Consortium (DKTK) Partner Site BerlinGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Roberto Arsie
- Max Delbrück Center for Molecular MedicineBerlin Institute for Medical Systems Biology (BIMSB)BerlinGermany
| | - Tzu‐Ting Wei
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Max Delbrück Center for Molecular MedicineBerlin Institute for Medical Systems Biology (BIMSB)BerlinGermany
| | - Kathleen Klotz‐Noack
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Institute of Medical ImmunologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Roland F Schwarz
- Max Delbrück Center for Molecular MedicineBerlin Institute for Medical Systems Biology (BIMSB)BerlinGermany
- BIFOLD – Berlin Institute for the Foundations of Learning and DataBerlinGermany
| | - Birgit Sawitzki
- Institute of Medical ImmunologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Carsten Kamphues
- German Cancer Consortium (DKTK) Partner Site BerlinGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Department of SurgeryCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Dieter Beule
- Core Unit Bioinformatics (CUBI)Berlin Institute of Health at Charité Universitätsmedizin – BerlinBerlinGermany
| | - Markus Landthaler
- Max Delbrück Center for Molecular MedicineBerlin Institute for Medical Systems Biology (BIMSB)BerlinGermany
| | - Christine Sers
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- German Cancer Consortium (DKTK) Partner Site BerlinGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - David Horst
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- German Cancer Consortium (DKTK) Partner Site BerlinGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Nils Blüthgen
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- IRI Life SciencesHumboldt University of BerlinBerlinGermany
- German Cancer Consortium (DKTK) Partner Site BerlinGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Markus Morkel
- Institute of PathologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- German Cancer Consortium (DKTK) Partner Site BerlinGerman Cancer Research Center (DKFZ)HeidelbergGermany
- BIH Bioportal Single CellsBerlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
| |
Collapse
|
64
|
Wang H, Yu S, Cai Q, Ma D, Yang L, Zhao J, Jiang L, Zhang X, Yu Z. The Prognostic Model Based on Tumor Cell Evolution Trajectory Reveals a Different Risk Group of Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:737723. [PMID: 34660596 PMCID: PMC8511531 DOI: 10.3389/fcell.2021.737723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide, and heterogeneity of HCC is the major barrier in improving patient outcome. To stratify HCC patients with different degrees of malignancy and provide precise treatment strategies, we reconstructed the tumor evolution trajectory with the help of scRNA-seq data and established a 30-gene prognostic model to identify the malignant state in HCC. Patients were divided into high-risk and low-risk groups. C-index and receiver operating characteristic (ROC) curve confirmed the excellent predictive value of this model. Downstream analysis revealed the underlying molecular and functional characteristics of this model, including significantly higher genomic instability and stronger proliferation/progression potential in the high-risk group. In summary, we established a novel prognostic model to overcome the barriers caused by HCC heterogeneity and provide the possibility of better clinical management for HCC patients to improve their survival outcomes.
Collapse
Affiliation(s)
- Haoren Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yunnan University, Kunming, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shizhe Yu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yunnan University, Kunming, China
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiang Cai
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Duo Ma
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Lingpeng Yang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Jian Zhao
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Long Jiang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Xinyi Zhang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Zhiyong Yu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yunnan University, Kunming, China
| |
Collapse
|
65
|
Dong J, Zhou P, Wu Y, Chen Y, Xie H, Gao Y, Lu J, Yang J, Zhang X, Wen L, Li T, Tang F. Integrating single-cell datasets with ambiguous batch information by incorporating molecular network features. Brief Bioinform 2021; 23:6373559. [PMID: 34553223 DOI: 10.1093/bib/bbab366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/30/2023] Open
Abstract
With the rapid development of single-cell sequencing techniques, several large-scale cell atlas projects have been launched across the world. However, it is still challenging to integrate single-cell RNA-seq (scRNA-seq) datasets with diverse tissue sources, developmental stages and/or few overlaps, due to the ambiguity in determining the batch information, which is particularly important for current batch-effect correction methods. Here, we present SCORE, a simple network-based integration methodology, which incorporates curated molecular network features to infer cellular states and generate a unified workflow for integrating scRNA-seq datasets. Validating on real single-cell datasets, we showed that regardless of batch information, SCORE outperforms existing methods in accuracy, robustness, scalability and data integration.
Collapse
Affiliation(s)
- Ji Dong
- Guangzhou Laboratory, Guangzhou, China
| | - Peijie Zhou
- Department of Mathematics, University of California at Irvine, Irvine, CA, USA
| | - Yichong Wu
- School of Mathematical Sciences, Peking University, Beijing, China
| | - Yidong Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Haoling Xie
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing, China
| | - Yuan Gao
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing, China
| | - Jiansen Lu
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing, China
| | - Jingwei Yang
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing, China
| | - Xiannian Zhang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lu Wen
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing, China
| | - Tiejun Li
- School of Mathematical Sciences, Peking University, Beijing, China
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
66
|
Stein CM, Weiskirchen R, Damm F, Strzelecka PM. Single-cell omics: Overview, analysis, and application in biomedical science. J Cell Biochem 2021; 122:1571-1578. [PMID: 34459502 DOI: 10.1002/jcb.30134] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/26/2021] [Accepted: 08/09/2021] [Indexed: 12/16/2022]
Abstract
Single-cell sequencing methods provide the highest resolution insight into cellular heterogeneity. Owing to their rapid growth and decreasing cost, they are now widely accessible to scientists worldwide. Single-cell technologies enable analysis of a large number of cells, making them powerful tools to characterise rare cell types and refine our understanding of diverse cell states. Moreover, single-cell application in biomedical sciences helps to unravel mechanisms related to disease pathogenesis and outcome. In this Viewpoint, we briefly describe existing single-cell methods (genomics, transcriptomics, epigenomics, proteomics, and mulitomics), comment on available analysis tools, and give examples of method applications in the biomedical field.
Collapse
Affiliation(s)
- Catarina M Stein
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin School Of Integrative Oncology (BSIO), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Frederik Damm
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Paulina M Strzelecka
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| |
Collapse
|
67
|
The genetic architecture of primary biliary cholangitis. Eur J Med Genet 2021; 64:104292. [PMID: 34303876 DOI: 10.1016/j.ejmg.2021.104292] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/03/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022]
Abstract
Primary biliary cholangitis (PBC) is a rare autoimmune disease of the liver affecting the small bile ducts. From a genetic point of view, PBC is a complex trait and several genetic and environmental factors have been called in action to explain its etiopathogenesis. Similarly to other complex traits, PBC has benefited from the introduction of genome-wide association studies (GWAS), which identified many variants predisposing or protecting toward the development of the disease. While a progressive endeavour toward the characterization of candidate loci and downstream pathways is currently ongoing, there is still a relatively large portion of heritability of PBC to be revealed. In addition, genetic variation behind progression of the disease and therapeutic response are mostly to be investigated yet. This review outlines the state-of-the-art regarding the genetic architecture of PBC and provides some hints for future investigations, focusing on the study of gene-gene interactions, the application of whole-genome sequencing techniques, and the investigation of X chromosome that can be helpful to cover the missing heritability gap in PBC.
Collapse
|
68
|
Abstract
Cell atlases are essential companions to the genome as they elucidate how genes are used in a cell type-specific manner or how the usage of genes changes over the lifetime of an organism. This review explores recent advances in whole-organism single-cell atlases, which enable understanding of cell heterogeneity and tissue and cell fate, both in health and disease. Here we provide an overview of recent efforts to build cell atlases across species and discuss the challenges that the field is currently facing. Moreover, we propose the concept of having a knowledgebase that can scale with the number of experiments and computational approaches and a new feedback loop for development and benchmarking of computational methods that includes contributions from the users. These two aspects are key for community efforts in single-cell biology that will help produce a comprehensive annotated map of cell types and states with unparalleled resolution.
Collapse
Affiliation(s)
| | - Bruno Tojo
- Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisbon, Portugal
| | - Aaron McGeever
- Chan Zuckerberg Biohub, San Francisco, California 94103, USA;
| |
Collapse
|
69
|
Zhao N, Rosen JM. Breast cancer heterogeneity through the lens of single-cell analysis and spatial pathologies. Semin Cancer Biol 2021; 82:3-10. [PMID: 34274486 DOI: 10.1016/j.semcancer.2021.07.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/30/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
Breast cancer ecosystems are composed of complex cell types, including tumor, stromal and immune cells, each of which can assume diverse phenotypes. Both the heterogeneous composition and spatially distinct tumor microenvironment impact breast cancer progression, treatment response and therapeutic resistance. Thus, a deeper understanding of breast cancer heterogeneity may help facilitate the development of novel therapies and improve outcomes for patients. The advent of paradigm shifting single-cell analysis and spatial pathologies allows for a comprehensive analysis of the tumor ecosystem as well as the interactions between its components at unprecedented resolution. In this review, we discuss the insights gained through single-cell analysis and spatial pathologies on breast cancer heterogeneity.
Collapse
Affiliation(s)
- Na Zhao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
70
|
Chiva-Blanch G, Mayr M. Lessons from the spatiotemporal expression patterns of RNA vs. proteins during the cell cycle. Cardiovasc Res 2021; 117:e91-e93. [PMID: 34179948 DOI: 10.1093/cvr/cvab196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Gemma Chiva-Blanch
- Endocrinology and Nutrition Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain.,Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
| | - Manuel Mayr
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, Strand, London WC2R 2LS, UK
| |
Collapse
|
71
|
Pinheiro I, Torres-Padilla ME, Almouzni G. Epigenomics in the single cell era, an important read out for genome function and cell identity. Epigenomics 2021; 13:981-984. [PMID: 34114476 DOI: 10.2217/epi-2021-0153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Inês Pinheiro
- Institut Curie, CNRS, PSL Research University, LabEx DEEP, Sorbonne Université, Nuclear Dynamics Unit, Equipe Labellisée Ligue Contre le Cancer, 75248 Paris Cedex 05, France
| | - Maria-Elena Torres-Padilla
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum München - German Research Center for Environmental Health, Munich 81377, Germany.,Faculty of Biology, Ludwig-Maximilians Universität, 82152 Martinsried, Germany
| | - Geneviève Almouzni
- Institut Curie, CNRS, PSL Research University, LabEx DEEP, Sorbonne Université, Nuclear Dynamics Unit, Equipe Labellisée Ligue Contre le Cancer, 75248 Paris Cedex 05, France
| |
Collapse
|
72
|
Liu J, Qu S, Zhang T, Gao Y, Shi H, Song K, Chen W, Yin W. Applications of Single-Cell Omics in Tumor Immunology. Front Immunol 2021; 12:697412. [PMID: 34177965 PMCID: PMC8221107 DOI: 10.3389/fimmu.2021.697412] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/17/2021] [Indexed: 12/20/2022] Open
Abstract
The tumor microenvironment (TME) is an ecosystem that contains various cell types, including cancer cells, immune cells, stromal cells, and many others. In the TME, cancer cells aggressively proliferate, evolve, transmigrate to the circulation system and other organs, and frequently communicate with adjacent immune cells to suppress local tumor immunity. It is essential to delineate this ecosystem's complex cellular compositions and their dynamic intercellular interactions to understand cancer biology and tumor immunology and to benefit tumor immunotherapy. But technically, this is extremely challenging due to the high complexities of the TME. The rapid developments of single-cell techniques provide us powerful means to systemically profile the multiple omics status of the TME at a single-cell resolution, shedding light on the pathogenic mechanisms of cancers and dysfunctions of tumor immunity in an unprecedently resolution. Furthermore, more advanced techniques have been developed to simultaneously characterize multi-omics and even spatial information at the single-cell level, helping us reveal the phenotypes and functionalities of disease-specific cell populations more comprehensively. Meanwhile, the connections between single-cell data and clinical characteristics are also intensively interrogated to achieve better clinical diagnosis and prognosis. In this review, we summarize recent progress in single-cell techniques, discuss their technical advantages, limitations, and applications, particularly in tumor biology and immunology, aiming to promote the research of cancer pathogenesis, clinically relevant cancer diagnosis, prognosis, and immunotherapy design with the help of single-cell techniques.
Collapse
Affiliation(s)
- Junwei Liu
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Saisi Qu
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Tongtong Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yufei Gao
- School of Mechanical Engineering, Zhejiang University, Hangzhou, China
| | - Hongyu Shi
- Department of Biological Testing, Zhejiang Puluoting Health Technology Co., Ltd., Hangzhou, China
| | - Kaichen Song
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Wei Chen
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, China
| | - Weiwei Yin
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
73
|
Marshall AS, Jones NS. Discovering Cellular Mitochondrial Heteroplasmy Heterogeneity with Single Cell RNA and ATAC Sequencing. BIOLOGY 2021; 10:503. [PMID: 34198745 PMCID: PMC8230039 DOI: 10.3390/biology10060503] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/14/2022]
Abstract
Next-generation sequencing technologies have revolutionised the study of biological systems by enabling the examination of a broad range of tissues. Its application to single-cell genomics has generated a dynamic and evolving field with a vast amount of research highlighting heterogeneity in transcriptional, genetic and epigenomic state between cells. However, compared to these aspects of cellular heterogeneity, relatively little has been gleaned from single-cell datasets regarding cellular mitochondrial heterogeneity. Single-cell sequencing techniques can provide coverage of the mitochondrial genome which allows researchers to probe heteroplasmies at the level of the single cell, and observe interactions with cellular function. In this review, we give an overview of two popular single-cell modalities-single-cell RNA sequencing and single-cell ATAC sequencing-whose throughput and widespread usage offers researchers the chance to probe heteroplasmy combined with cell state in detailed resolution across thousands of cells. After summarising these technologies in the context of mitochondrial research, we give an overview of recent methods which have used these approaches for discovering mitochondrial heterogeneity. We conclude by highlighting current limitations of these approaches and open problems for future consideration.
Collapse
Affiliation(s)
| | - Nick S. Jones
- Department of Mathematics, Imperial College London, Huxley Building, South Kensington Campus, London SW7 2AZ, UK;
| |
Collapse
|
74
|
Rizzo G, Bertotti A, Leto SM, Vetrano S. Patient-derived tumor models: a more suitable tool for pre-clinical studies in colorectal cancer. J Exp Clin Cancer Res 2021; 40:178. [PMID: 34074330 PMCID: PMC8168319 DOI: 10.1186/s13046-021-01970-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/02/2021] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer (CRC), despite the advances in screening and surveillance, remains the second most common cause of cancer death worldwide. The biological inadequacy of pre-clinical models to fully recapitulate the multifactorial etiology and the complexity of tumor microenvironment and human CRC's genetic heterogeneity has limited cancer treatment development. This has led to the development of Patient-derived models able to phenocopy as much as possible the original inter- and intra-tumor heterogeneity of CRC, reflecting the tumor microenvironment's cellular interactions. Implantation of patient tissue into immunodeficient mice hosts and the culture of tumor organoids have allowed advances in cancer biology and metastasis. This review highlights the advantages and limits of Patient-derived models as innovative and valuable pre-clinical tools to study progression and metastasis of CRC, develop novel therapeutic strategies by creating a drug screening platform, and predict the efficacy of clinical response to therapy.
Collapse
Affiliation(s)
- Giulia Rizzo
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, 20090, Milan, Italy
| | - Andrea Bertotti
- Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute - FPO IRCCs, Candiolo, 10060, Torino, Italy
- Department of Oncology, University of Torino School of Medicine, Candiolo, 10060, Torino, Italy
| | - Simonetta Maria Leto
- Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute - FPO IRCCs, Candiolo, 10060, Torino, Italy
| | - Stefania Vetrano
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, 20090, Milan, Italy.
- IBD Center, Department of Gastroenterology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy.
| |
Collapse
|
75
|
Opportunities and challenges for microRNA-targeting therapeutics for epilepsy. Trends Pharmacol Sci 2021; 42:605-616. [PMID: 33992468 DOI: 10.1016/j.tips.2021.04.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/30/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022]
Abstract
Epilepsy is a common and serious neurological disorder characterised by recurrent spontaneous seizures. Frontline pharmacotherapy includes small-molecule antiseizure drugs that typically target ion channels and neurotransmitter systems, but these fail in 30% of patients and do not prevent either the development or progression of epilepsy. An emerging therapeutic target is microRNA (miRNA), small noncoding RNAs that negatively regulate sets of proteins. Their multitargeting action offers unique advantages for certain forms of epilepsy with complex underlying pathophysiology, such as temporal lobe epilepsy (TLE). miRNA can be inhibited by designed antisense oligonucleotides (ASOs; e.g., antimiRs). Here, we outline the prospects for miRNA-based therapies. We review design considerations for nucleic acid-based approaches and the challenges and next steps in developing therapeutic miRNA-targeting molecules for epilepsy.
Collapse
|
76
|
Computational principles and challenges in single-cell data integration. Nat Biotechnol 2021; 39:1202-1215. [PMID: 33941931 DOI: 10.1038/s41587-021-00895-7] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
The development of single-cell multimodal assays provides a powerful tool for investigating multiple dimensions of cellular heterogeneity, enabling new insights into development, tissue homeostasis and disease. A key challenge in the analysis of single-cell multimodal data is to devise appropriate strategies for tying together data across different modalities. The term 'data integration' has been used to describe this task, encompassing a broad collection of approaches ranging from batch correction of individual omics datasets to association of chromatin accessibility and genetic variation with transcription. Although existing integration strategies exploit similar mathematical ideas, they typically have distinct goals and rely on different principles and assumptions. Consequently, new definitions and concepts are needed to contextualize existing methods and to enable development of new methods.
Collapse
|
77
|
Bayesian inference of gene expression states from single-cell RNA-seq data. Nat Biotechnol 2021; 39:1008-1016. [PMID: 33927416 DOI: 10.1038/s41587-021-00875-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 02/26/2021] [Indexed: 12/14/2022]
Abstract
Despite substantial progress in single-cell RNA-seq (scRNA-seq) data analysis methods, there is still little agreement on how to best normalize such data. Starting from the basic requirements that inferred expression states should correct for both biological and measurement sampling noise and that changes in expression should be measured in terms of fold changes, we here derive a Bayesian normalization procedure called Sanity (SAmpling-Noise-corrected Inference of Transcription activitY) from first principles. Sanity estimates expression values and associated error bars directly from raw unique molecular identifier (UMI) counts without any tunable parameters. Using simulated and real scRNA-seq datasets, we show that Sanity outperforms other normalization methods on downstream tasks, such as finding nearest-neighbor cells and clustering cells into subtypes. Moreover, we show that by systematically overestimating the expression variability of genes with low expression and by introducing spurious correlations through mapping the data to a lower-dimensional representation, other methods yield severely distorted pictures of the data.
Collapse
|
78
|
Mayr CH, Simon LM, Leuschner G, Ansari M, Schniering J, Geyer PE, Angelidis I, Strunz M, Singh P, Kneidinger N, Reichenberger F, Silbernagel E, Böhm S, Adler H, Lindner M, Maurer B, Hilgendorff A, Prasse A, Behr J, Mann M, Eickelberg O, Theis FJ, Schiller HB. Integrative analysis of cell state changes in lung fibrosis with peripheral protein biomarkers. EMBO Mol Med 2021; 13:e12871. [PMID: 33650774 PMCID: PMC8033531 DOI: 10.15252/emmm.202012871] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/05/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
The correspondence of cell state changes in diseased organs to peripheral protein signatures is currently unknown. Here, we generated and integrated single-cell transcriptomic and proteomic data from multiple large pulmonary fibrosis patient cohorts. Integration of 233,638 single-cell transcriptomes (n = 61) across three independent cohorts enabled us to derive shifts in cell type proportions and a robust core set of genes altered in lung fibrosis for 45 cell types. Mass spectrometry analysis of lung lavage fluid (n = 124) and plasma (n = 141) proteomes identified distinct protein signatures correlated with diagnosis, lung function, and injury status. A novel SSTR2+ pericyte state correlated with disease severity and was reflected in lavage fluid by increased levels of the complement regulatory factor CFHR1. We further discovered CRTAC1 as a biomarker of alveolar type-2 epithelial cell health status in lavage fluid and plasma. Using cross-modal analysis and machine learning, we identified the cellular source of biomarkers and demonstrated that information transfer between modalities correctly predicts disease status, suggesting feasibility of clinical cell state monitoring through longitudinal sampling of body fluid proteomes.
Collapse
Affiliation(s)
- Christoph H Mayr
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Lukas M Simon
- Institute of Computational BiologyHelmholtz Zentrum MünchenMunichGermany
| | - Gabriela Leuschner
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
- Department of Internal Medicine VLudwig‐Maximilians University (LMU) MunichMember of the German Center for Lung Research (DZL), CPC‐M bioArchiveMunichGermany
| | - Meshal Ansari
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
- Institute of Computational BiologyHelmholtz Zentrum MünchenMunichGermany
| | - Janine Schniering
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
- Department of RheumatologyCenter of Experimental RheumatologyUniversity & University Hospital ZurichZurichSwitzerland
| | - Philipp E Geyer
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Ilias Angelidis
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Maximilian Strunz
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Pawandeep Singh
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Nikolaus Kneidinger
- Department of Internal Medicine VLudwig‐Maximilians University (LMU) MunichMember of the German Center for Lung Research (DZL), CPC‐M bioArchiveMunichGermany
| | - Frank Reichenberger
- Asklepios Fachkliniken Munich‐GautingCPC‐M bioArchive, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Edith Silbernagel
- Asklepios Fachkliniken Munich‐GautingCPC‐M bioArchive, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Stephan Böhm
- Faculty of MedicineMax von Pettenkofer‐Institute, VirologyNational Reference Center for RetrovirusesLMU MünchenMunichGermany
| | - Heiko Adler
- Helmholtz Zentrum MünchenResearch Unit Lung Repair and Regeneration, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Michael Lindner
- Asklepios Fachkliniken Munich‐GautingCPC‐M bioArchive, Member of the German Center for Lung Research (DZL)MunichGermany
- University Department of Visceral and Thoracic Surgery SalzburgParacelsus Medical UniversitySalzburgAustria
| | - Britta Maurer
- Department of RheumatologyCenter of Experimental RheumatologyUniversity & University Hospital ZurichZurichSwitzerland
| | - Anne Hilgendorff
- Center for Comprehensive Developmental Care (CDeCLMU)Member of the German Center for Lung Research (DZL)Hospital of the Ludwig‐Maximilians University (LMU)CPC‐M bioArchiveMunichGermany
| | - Antje Prasse
- Department of PneumologyHannover Medical School, Member of the German Center for Lung Research (DZL)HannoverGermany
| | - Jürgen Behr
- Department of Internal Medicine VLudwig‐Maximilians University (LMU) MunichMember of the German Center for Lung Research (DZL), CPC‐M bioArchiveMunichGermany
- Asklepios Fachkliniken Munich‐GautingCPC‐M bioArchive, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Matthias Mann
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy, and Critical Care MedicineDepartment of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Fabian J Theis
- Institute of Computational BiologyHelmholtz Zentrum MünchenMunichGermany
| | - Herbert B Schiller
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
| |
Collapse
|
79
|
Cirillo D, Núñez‐Carpintero I, Valencia A. Artificial intelligence in cancer research: learning at different levels of data granularity. Mol Oncol 2021; 15:817-829. [PMID: 33533192 PMCID: PMC8024732 DOI: 10.1002/1878-0261.12920] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/20/2020] [Accepted: 01/10/2021] [Indexed: 02/06/2023] Open
Abstract
From genome-scale experimental studies to imaging data, behavioral footprints, and longitudinal healthcare records, the convergence of big data in cancer research and the advances in Artificial Intelligence (AI) is paving the way to develop a systems view of cancer. Nevertheless, this biomedical area is largely characterized by the co-existence of big data and small data resources, highlighting the need for a deeper investigation about the crosstalk between different levels of data granularity, including varied sample sizes, labels, data types, and other data descriptors. This review introduces the current challenges, limitations, and solutions of AI in the heterogeneous landscape of data granularity in cancer research. Such a variety of cancer molecular and clinical data calls for advancing the interoperability among AI approaches, with particular emphasis on the synergy between discriminative and generative models that we discuss in this work with several examples of techniques and applications.
Collapse
Affiliation(s)
| | | | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC)BarcelonaSpain
- ICREABarcelonaSpain
| |
Collapse
|
80
|
Schultze JL, Aschenbrenner AC. COVID-19 and the human innate immune system. Cell 2021; 184:1671-1692. [PMID: 33743212 PMCID: PMC7885626 DOI: 10.1016/j.cell.2021.02.029] [Citation(s) in RCA: 443] [Impact Index Per Article: 147.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/19/2021] [Accepted: 02/10/2021] [Indexed: 01/08/2023]
Abstract
The introduction of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into the human population represents a tremendous medical and economic crisis. Innate immunity-as the first line of defense of our immune system-plays a central role in combating this novel virus. Here, we provide a conceptual framework for the interaction of the human innate immune system with SARS-CoV-2 to link the clinical observations with experimental findings that have been made during the first year of the pandemic. We review evidence that variability in innate immune system components among humans is a main contributor to the heterogeneous disease courses observed for coronavirus disease 2019 (COVID-19), the disease spectrum induced by SARS-CoV-2. A better understanding of the pathophysiological mechanisms observed for cells and soluble mediators involved in innate immunity is a prerequisite for the development of diagnostic markers and therapeutic strategies targeting COVID-19. However, this will also require additional studies addressing causality of events, which so far are lagging behind.
Collapse
Affiliation(s)
- Joachim L Schultze
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; PRECISE Platform for Single Cell Genomics and Epigenomics at the DZNE and the University of Bonn, Bonn, Germany; Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.
| | - Anna C Aschenbrenner
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; PRECISE Platform for Single Cell Genomics and Epigenomics at the DZNE and the University of Bonn, Bonn, Germany; Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
81
|
Hoffmann A, Spengler D. Single-Cell Transcriptomics Supports a Role of CHD8 in Autism. Int J Mol Sci 2021; 22:3261. [PMID: 33806835 PMCID: PMC8004931 DOI: 10.3390/ijms22063261] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/16/2021] [Accepted: 03/20/2021] [Indexed: 12/16/2022] Open
Abstract
Chromodomain helicase domain 8 (CHD8) is one of the most frequently mutated and most penetrant genes in the autism spectrum disorder (ASD). Individuals with CHD8 mutations show leading symptoms of autism, macrocephaly, and facial dysmorphisms. The molecular and cellular mechanisms underpinning the early onset and development of these symptoms are still poorly understood and prevent timely and more efficient therapies of patients. Progress in this area will require an understanding of "when, why and how cells deviate from their normal trajectories". High-throughput single-cell RNA sequencing (sc-RNAseq) directly quantifies information-bearing RNA molecules that enact each cell's biological identity. Here, we discuss recent insights from sc-RNAseq of CRISPR/Cas9-editing of Chd8/CHD8 during mouse neocorticogenesis and human cerebral organoids. Given that the deregulation of the balance between excitation and inhibition (E/I balance) in cortical and subcortical circuits is thought to represent a major etiopathogenetic mechanism in ASD, we focus on the question of whether, and to what degree, results from current sc-RNAseq studies support this hypothesis. Beyond that, we discuss the pros and cons of these approaches and further steps to be taken to harvest the full potential of these transformative techniques.
Collapse
Affiliation(s)
| | - Dietmar Spengler
- Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany;
| |
Collapse
|
82
|
Irmisch A, Bonilla X, Chevrier S, Lehmann KV, Singer F, Toussaint NC, Esposito C, Mena J, Milani ES, Casanova R, Stekhoven DJ, Wegmann R, Jacob F, Sobottka B, Goetze S, Kuipers J, Sarabia Del Castillo J, Prummer M, Tuncel MA, Menzel U, Jacobs A, Engler S, Sivapatham S, Frei AL, Gut G, Ficek J, Miglino N, Aebersold R, Bacac M, Beerenwinkel N, Beisel C, Bodenmiller B, Dummer R, Heinzelmann-Schwarz V, Koelzer VH, Manz MG, Moch H, Pelkmans L, Snijder B, Theocharides APA, Tolnay M, Wicki A, Wollscheid B, Rätsch G, Levesque MP. The Tumor Profiler Study: integrated, multi-omic, functional tumor profiling for clinical decision support. Cancer Cell 2021; 39:288-293. [PMID: 33482122 DOI: 10.1016/j.ccell.2021.01.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The application and integration of molecular profiling technologies create novel opportunities for personalized medicine. Here, we introduce the Tumor Profiler Study, an observational trial combining a prospective diagnostic approach to assess the relevance of in-depth tumor profiling to support clinical decision-making with an exploratory approach to improve the biological understanding of the disease.
Collapse
Affiliation(s)
- Anja Irmisch
- University Hospital Zurich, Department of Dermatology, University of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland
| | - Ximena Bonilla
- ETH Zurich, Department of Computer Science, Institute of Machine Learning, Universitätstrasse 6, 8092 Zurich, Switzerland; SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Hospital Zurich, Biomedical Informatics, Schmelzbergstrasse 26, 8006 Zurich, Switzerland
| | - Stéphane Chevrier
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Kjong-Van Lehmann
- ETH Zurich, Department of Computer Science, Institute of Machine Learning, Universitätstrasse 6, 8092 Zurich, Switzerland; SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Hospital Zurich, Biomedical Informatics, Schmelzbergstrasse 26, 8006 Zurich, Switzerland
| | - Franziska Singer
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, NEXUS Personalized Health Technologies, John-von-Neumann-Weg 9, 8093 Zurich, Switzerland
| | - Nora C Toussaint
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, NEXUS Personalized Health Technologies, John-von-Neumann-Weg 9, 8093 Zurich, Switzerland
| | - Cinzia Esposito
- University of Zurich, Department of Molecular Life Sciences, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Julien Mena
- ETH Zurich, Department of Biology, Institute of Molecular Systems Biology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Emanuela S Milani
- ETH Zurich, Department of Health Sciences and Technology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Ruben Casanova
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Daniel J Stekhoven
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, NEXUS Personalized Health Technologies, John-von-Neumann-Weg 9, 8093 Zurich, Switzerland
| | - Rebekka Wegmann
- ETH Zurich, Department of Biology, Institute of Molecular Systems Biology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Francis Jacob
- University Hospital Basel and University of Basel, Department of Biomedicine, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Bettina Sobottka
- University Hospital Zurich, Department of Pathology and Molecular Pathology, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Sandra Goetze
- ETH Zurich, Department of Health Sciences and Technology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Jack Kuipers
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Jacobo Sarabia Del Castillo
- University of Zurich, Department of Molecular Life Sciences, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Michael Prummer
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, NEXUS Personalized Health Technologies, John-von-Neumann-Weg 9, 8093 Zurich, Switzerland
| | - Mustafa A Tuncel
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Ulrike Menzel
- ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Andrea Jacobs
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Stefanie Engler
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Sujana Sivapatham
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Anja L Frei
- University Hospital Zurich, Department of Pathology and Molecular Pathology, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Gabriele Gut
- University of Zurich, Department of Molecular Life Sciences, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Joanna Ficek
- ETH Zurich, Department of Computer Science, Institute of Machine Learning, Universitätstrasse 6, 8092 Zurich, Switzerland; SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Hospital Zurich, Biomedical Informatics, Schmelzbergstrasse 26, 8006 Zurich, Switzerland
| | - Nicola Miglino
- University Hospital Zurich, Department of Medical Oncology and Hematology, Rämistrasse 100, 8091 Zurich, Switzerland
| | | | - Rudolf Aebersold
- ETH Zurich, Department of Biology, Institute of Molecular Systems Biology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Marina Bacac
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Niko Beerenwinkel
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Christian Beisel
- ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Bernd Bodenmiller
- University of Zurich, Department of Quantitative Biomedicine, Winterthurerstrasse 190, 8057 Zurich, Switzerland; ETH Zurich, Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Reinhard Dummer
- University Hospital Zurich, Department of Dermatology, University of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland
| | - Viola Heinzelmann-Schwarz
- University Hospital Basel and University of Basel, Department of Biomedicine, Hebelstrasse 20, 4031 Basel, Switzerland; University Hospital Basel, Gynecological Cancer Center, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Viktor H Koelzer
- University Hospital Zurich, Department of Pathology and Molecular Pathology, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Markus G Manz
- University Hospital Zurich, Department of Medical Oncology and Hematology, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Holger Moch
- University Hospital Zurich, Department of Pathology and Molecular Pathology, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Lucas Pelkmans
- University of Zurich, Department of Molecular Life Sciences, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Berend Snijder
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; ETH Zurich, Department of Biology, Institute of Molecular Systems Biology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Alexandre P A Theocharides
- University Hospital Zurich, Department of Medical Oncology and Hematology, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Markus Tolnay
- University Hospital Basel, Institute of Medical Genetics and Pathology, Schönbeinstrasse 40, 4031 Basel, Switzerland
| | - Andreas Wicki
- University Hospital Zurich, Department of Medical Oncology and Hematology, Rämistrasse 100, 8091 Zurich, Switzerland; University of Zurich, Faculty of Medicine, Zurich, Switzerland
| | - Bernd Wollscheid
- ETH Zurich, Department of Health Sciences and Technology, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Gunnar Rätsch
- ETH Zurich, Department of Computer Science, Institute of Machine Learning, Universitätstrasse 6, 8092 Zurich, Switzerland; SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Hospital Zurich, Biomedical Informatics, Schmelzbergstrasse 26, 8006 Zurich, Switzerland; ETH Zurich, Department of Biology, Wolfgang-Pauli-Strasse 27, 8093 Zurich, Switzerland.
| | - Mitchell P Levesque
- University Hospital Zurich, Department of Dermatology, University of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland.
| |
Collapse
|
83
|
Wiltfang J, Esselmann H, Barnikol UB. [The Use of Artificial Intelligence in Alzheimer's Disease - Personalized Diagnostics and Therapy]. PSYCHIATRISCHE PRAXIS 2021; 48:S31-S36. [PMID: 33652485 DOI: 10.1055/a-1369-3133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Using the example of dementia in Alzheimer's disease, it is shown which opportunities but also risks are posed by newer methodological approaches of artificial intelligence (AI) for the diagnosis and treatment of Alzheimer's dementia (AD). In addition, AI is examined in the context of an ethical-philosophical critique of technology.
Collapse
Affiliation(s)
- Jens Wiltfang
- Klinik für Psychiatrie und Psychotherapie, Universitätsmedizin Göttingen.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Standort Göttingen (DZNE-Göttingen)
| | - Hermann Esselmann
- Klinik für Psychiatrie und Psychotherapie, Universitätsmedizin Göttingen
| | - Utako B Barnikol
- Angewandte Ethik in der translationalen Krebsforschung, Clearingstelle Ethik, Centrum für Integrierte Onkologie (CIO), Uniklinik Köln
| |
Collapse
|
84
|
Translational insights from single-cell technologies across the cardiovascular disease continuum. Trends Cardiovasc Med 2021; 32:127-135. [PMID: 33667644 DOI: 10.1016/j.tcm.2021.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide. The societal health burden it represents can be reduced by taking preventive measures and developing more effective therapies. Reaching these goals, however, requires a better understanding of the pathophysiological processes leading to and occurring in the diseased heart. In the last 5 years, several biological advances applying single-cell technologies have enabled researchers to study cardiovascular diseases with unprecedented resolution. This has produced many new insights into how specific cell types change their gene expression level, activation status and potential cellular interactions with the development of cardiovascular disease, but a comprehensive overview of the clinical implications of these findings is lacking. In this review, we summarize and discuss these recent advances and the promise of single-cell technologies from a translational perspective across the cardiovascular disease continuum, covering both animal and human studies, and explore the future directions of the field.
Collapse
|
85
|
Aslan JE. Platelet Proteomes, Pathways, and Phenotypes as Informants of Vascular Wellness and Disease. Arterioscler Thromb Vasc Biol 2021; 41:999-1011. [PMID: 33441027 PMCID: PMC7980774 DOI: 10.1161/atvbaha.120.314647] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platelets rapidly undergo responsive transitions in form and function to repair vascular endothelium and mediate hemostasis. In contrast, heterogeneous platelet subpopulations with a range of primed or refractory phenotypes gradually arise in chronic inflammatory and other conditions in a manner that may indicate or support disease. Qualitatively distinguishable platelet phenotypes are increasingly associated with a variety of physiological and pathological circumstances; however, the origins and significance of platelet phenotypic variation remain unclear and conceptually vague. As changes in platelet function in disease exhibit many similarities to platelets following the activation of platelet agonist receptors, the intracellular responses of platelets common to hemostasis and inflammation may provide insights to the molecular basis of platelet phenotype. Here, we review concepts around how protein-level relations-from platelet receptors through intracellular signaling events-may help to define platelet phenotypes in inflammation, immune responses, aging, and other conditions. We further discuss how representing systems-wide platelet proteomics data profiles as circuit-like networks of causally related intracellular events, or, pathway maps, may inform molecular definitions of platelet phenotype. In addition to offering insights into platelets as druggable targets, maps of causally arranged intracellular relations underlying platelet function can also advance precision and interceptive medicine efforts by leveraging platelets as accessible, dynamic, endogenous, circulating biomarkers of vascular wellness and disease. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Joseph E. Aslan
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Chemical Physiology and Biochemistry and School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
86
|
Yamada S, Behfar A, Terzic A. Regenerative medicine clinical readiness. Regen Med 2021; 16:309-322. [PMID: 33622049 PMCID: PMC8050983 DOI: 10.2217/rme-2020-0178] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
Regenerative medicine, poised to transform 21st century healthcare, has aspired to enrich care options by bringing cures to patients in need. Science-driven responsible and regulated translation of innovative technology has enabled the launch of previously unimaginable care pathways adopted prudently for select serious diseases and disabilities. The collective resolve to advance the design, manufacture and validity of affordable regenerative solutions aims to democratize such health benefits for all. The objective of this Review is to outline the framework and prerequisites that underpin clinical readiness of regenerative care. Integrated research and development, specialized workforce education and accessible evidence-based practice implementation are at the core of realizing an equitable regenerative medicine vision.
Collapse
Affiliation(s)
- Satsuki Yamada
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, 55905 MN, USA
- Division of Geriatric Medicine & Gerontology, Department of Medicine, Mayo Clinic, Rochester, 55905 MN, USA
| | - Atta Behfar
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, 55905 MN, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, 55905 MN, USA
| | - Andre Terzic
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, 55905 MN, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Department of Clinical Genomics, Mayo Clinic, Rochester, 55905 MN, USA
| |
Collapse
|
87
|
Aerts L, Lauwers E, De Strooper B. Dementia and COVID-19: a health and research funding crisis. Lancet Neurol 2021; 20:90. [PMID: 33484651 DOI: 10.1016/s1474-4422(21)00002-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/04/2021] [Indexed: 12/23/2022]
Affiliation(s)
- Liesbeth Aerts
- VIB-KU Leuven Center for Brain and Disease Research, and Department of Neurosciences, Leuven Brain Institute, KU Leuven, Belgium
| | - Elsa Lauwers
- VIB-KU Leuven Center for Brain and Disease Research, and Department of Neurosciences, Leuven Brain Institute, KU Leuven, Belgium
| | - Bart De Strooper
- VIB-KU Leuven Center for Brain and Disease Research, and Department of Neurosciences, Leuven Brain Institute, KU Leuven, Belgium; UK Dementia Research Institute, University College London WC1E 6BT, London, UK.
| |
Collapse
|
88
|
Caporale N, Testa G. COVID-19 lessons from the dish: Dissecting CNS manifestations through brain organoids. EMBO J 2021; 40:e107213. [PMID: 33175425 PMCID: PMC7809698 DOI: 10.15252/embj.2020107213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 11/10/2020] [Indexed: 01/07/2023] Open
Abstract
COVID-19 is increasingly understood as a systemic disease with pathogenic manifestations beyond the respiratory tract. Recent work by Ramani et al (2020) dissects the cellular and molecular mechanisms of SARS-CoV-2's neurotrophic properties, using viral exposure of human brain organoids. Their findings highlight neurons as primary target of cerebral SARS-CoV-2 infection and uncover its Tau-related neurotoxicity.
Collapse
Affiliation(s)
- Nicolò Caporale
- Department of Experimental OncologyIEOEuropean Institute of OncologyIRCCSMilanItaly
- Department of Oncology and Hemato‐oncologyUniversity of MilanMilanItaly
- Human TechnopoleMilanItaly
| | - Giuseppe Testa
- Department of Experimental OncologyIEOEuropean Institute of OncologyIRCCSMilanItaly
- Department of Oncology and Hemato‐oncologyUniversity of MilanMilanItaly
- Human TechnopoleMilanItaly
| |
Collapse
|
89
|
Single-cell RNA sequencing reveals distinct tumor microenvironmental patterns in lung adenocarcinoma. Oncogene 2021; 40:6748-6758. [PMID: 34663877 PMCID: PMC8677623 DOI: 10.1038/s41388-021-02054-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/16/2021] [Accepted: 09/30/2021] [Indexed: 12/24/2022]
Abstract
Recent developments in immuno-oncology demonstrate that not only cancer cells, but also the tumor microenvironment can guide precision medicine. A comprehensive and in-depth characterization of the tumor microenvironment is challenging since its cell populations are diverse and can be important even if scarce. To identify clinically relevant microenvironmental and cancer features, we applied single-cell RNA sequencing to ten human lung adenocarcinomas and ten normal control tissues. Our analyses revealed heterogeneous carcinoma cell transcriptomes reflecting histological grade and oncogenic pathway activities, and two distinct microenvironmental patterns. The immune-activated CP²E microenvironment was composed of cancer-associated myofibroblasts, proinflammatory monocyte-derived macrophages, plasmacytoid dendritic cells and exhausted CD8+ T cells, and was prognostically unfavorable. In contrast, the inert N³MC microenvironment was characterized by normal-like myofibroblasts, non-inflammatory monocyte-derived macrophages, NK cells, myeloid dendritic cells and conventional T cells, and was associated with a favorable prognosis. Microenvironmental marker genes and signatures identified in single-cell profiles had progonostic value in bulk tumor profiles. In summary, single-cell RNA profiling of lung adenocarcinoma provides additional prognostic information based on the microenvironment, and may help to predict therapy response and to reveal possible target cell populations for future therapeutic approaches.
Collapse
|
90
|
Tran F, Klein C, Arlt A, Imm S, Knappe E, Simmons A, Rosenstiel P, Seibler P. Stem Cells and Organoid Technology in Precision Medicine in Inflammation: Are We There Yet? Front Immunol 2020; 11:573562. [PMID: 33408713 PMCID: PMC7779798 DOI: 10.3389/fimmu.2020.573562] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Individualised cellular models of disease are a key tool for precision medicine to recapitulate chronic inflammatory processes. Organoid models can be derived from induced pluripotent stem cells (iPSCs) or from primary stem cells ex vivo. These models have been emerging over the past decade and have been used to reconstruct the respective organ-specific physiology and pathology, at an unsurpassed depth. In cancer research, patient-derived cancer organoids opened new perspectives in predicting therapy response and provided novel insights into tumour biology. In precision medicine of chronic inflammatory disorders, stem-cell based organoid models are currently being evaluated in pre-clinical pharmacodynamic studies (clinical studies in a dish) and are employed in clinical studies, e.g., by re-transplanting autologous epithelial organoids to re-establish intestinal barrier integrity. A particularly exciting feature of iPSC systems is their ability to provide insights into organ systems and inflammatory disease processes, which cannot be monitored with clinical biopsies, such as immune reactions in neurodegenerative disorders. Refinement of differentiation protocols, and next-generation co-culturing methods, aimed at generating self-organised, complex tissues in vitro, will be the next logical steps. In this mini-review, we critically discuss the current state-of-the-art stem cell and organoid technologies, as well as their future impact, potential and promises in combating immune-mediated chronic diseases.
Collapse
Affiliation(s)
- Florian Tran
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany.,Klinik für Innere Medizin I, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Alexander Arlt
- Klinik für Innere Medizin I, Universitätsklinikum Schleswig-Holstein, Kiel, Germany.,University Department for Gastroenterology, Klinikum Oldenburg AöR, European Medical School (EMS), Oldenburg, Germany
| | - Simon Imm
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Evelyn Knappe
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Alison Simmons
- MRC Human Immunology Unit (MRC), University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
91
|
Uyar B, Palmer D, Kowald A, Murua Escobar H, Barrantes I, Möller S, Akalin A, Fuellen G. Single-cell analyses of aging, inflammation and senescence. Ageing Res Rev 2020; 64:101156. [PMID: 32949770 PMCID: PMC7493798 DOI: 10.1016/j.arr.2020.101156] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 02/09/2023]
Abstract
Single-cell gene expression (transcriptomics) data are becoming robust and abundant, and are increasingly used to track organisms along their life-course. This allows investigation into how aging affects cellular transcriptomes, and how changes in transcriptomes may underlie aging, including chronic inflammation (inflammaging), immunosenescence and cellular senescence. We compiled and tabulated aging-related single-cell datasets published to date, collected and discussed relevant findings, and inspected some of these datasets ourselves. We specifically note insights that cannot (or not easily) be based on bulk data. For example, in some datasets, the fraction of cells expressing p16 (CDKN2A), one of the most prominent markers of cellular senescence, was reported to increase, in addition to its upregulated mean expression over all cells. Moreover, we found evidence for inflammatory processes in most datasets, some of these driven by specific cells of the immune system. Further, single-cell data are specifically useful to investigate whether transcriptional heterogeneity (also called noise or variability) increases with age, and many (but not all) studies in our review report an increase in such heterogeneity. Finally, we demonstrate some stability of marker gene expression patterns across closely similar studies and suggest that single-cell experiments may hold the key to provide detailed insights whenever interventions (countering aging, inflammation, senescence, disease, etc.) are affecting cells depending on cell type.
Collapse
Affiliation(s)
- Bora Uyar
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Daniel Palmer
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Axel Kowald
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Hugo Murua Escobar
- Rostock University Medical Center, Department of Hematology, Oncology and Palliative Medicine, Department of Medicine III, Rostock, Germany
| | - Israel Barrantes
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Steffen Möller
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Altuna Akalin
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Georg Fuellen
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany.
| |
Collapse
|
92
|
Imdahl F, Saliba AE. Advances and challenges in single-cell RNA-seq of microbial communities. Curr Opin Microbiol 2020; 57:102-110. [PMID: 33160164 DOI: 10.1016/j.mib.2020.10.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 09/16/2020] [Accepted: 10/02/2020] [Indexed: 12/17/2022]
Abstract
Microbes have developed complex strategies to respond to their environment and escape the immune system by individualizing their behavior. While single-cell RNA sequencing has become instrumental for studying mammalian cells, its use with fungi, protozoa and bacteria is still in its infancy. In this review, we highlight the major progress towards mapping the molecular states of microbes at the single-cell level using genome-wide transcriptomics and describe how these technologies can be extended to probe thousands of species at high throughput. We envision that mammalian and microbial single-cell profiling could soon be integrated for the study of microbial communities in health and disease.
Collapse
Affiliation(s)
- Fabian Imdahl
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany.
| |
Collapse
|
93
|
Partel G, Hilscher MM, Milli G, Solorzano L, Klemm AH, Nilsson M, Wählby C. Automated identification of the mouse brain's spatial compartments from in situ sequencing data. BMC Biol 2020; 18:144. [PMID: 33076915 PMCID: PMC7574211 DOI: 10.1186/s12915-020-00874-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 09/18/2020] [Indexed: 01/03/2023] Open
Abstract
Background Neuroanatomical compartments of the mouse brain are identified and outlined mainly based on manual annotations of samples using features related to tissue and cellular morphology, taking advantage of publicly available reference atlases. However, this task is challenging since sliced tissue sections are rarely perfectly parallel or angled with respect to sections in the reference atlas and organs from different individuals may vary in size and shape and requires manual annotation. With the advent of in situ sequencing technologies and automated approaches, it is now possible to profile the gene expression of targeted genes inside preserved tissue samples and thus spatially map biological processes across anatomical compartments. Results Here, we show how in situ sequencing data combined with dimensionality reduction and clustering can be used to identify spatial compartments that correspond to known anatomical compartments of the brain. We also visualize gradients in gene expression and sharp as well as smooth transitions between different compartments. We apply our method on mouse brain sections and show that a fully unsupervised approach can computationally define anatomical compartments, which are highly reproducible across individuals, using as few as 18 gene markers. We also show that morphological variation does not always follow gene expression, and different spatial compartments can be defined by various cell types with common morphological features but distinct gene expression profiles. Conclusion We show that spatial gene expression data can be used for unsupervised and unbiased annotations of mouse brain spatial compartments based only on molecular markers, without the need of subjective manual annotations based on tissue and cell morphology or matching reference atlases.
Collapse
Affiliation(s)
- Gabriele Partel
- Centre for Image Analysis, Department of Information Technology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Markus M Hilscher
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Giorgia Milli
- Centre for Image Analysis, Department of Information Technology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Leslie Solorzano
- Centre for Image Analysis, Department of Information Technology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anna H Klemm
- Centre for Image Analysis, Department of Information Technology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,BioImage Informatics Facility of SciLifeLab, Uppsala, Sweden
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Carolina Wählby
- Centre for Image Analysis, Department of Information Technology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden. .,BioImage Informatics Facility of SciLifeLab, Uppsala, Sweden.
| |
Collapse
|
94
|
Torres‐Padilla M, Bredenoord AL, Jongsma KR, Lunkes A, Marelli L, Pinheiro I, Testa G. Thinking "ethical" when designing an international, cross-disciplinary biomedical research consortium. EMBO J 2020; 39:e105725. [PMID: 32894572 PMCID: PMC7527923 DOI: 10.15252/embj.2020105725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This commentary outlines challenges with identifying and implementing ethical, legal and societal considerations when initiating large-scale scientific programs and suggests best practices to ensure responsible research.
Collapse
Affiliation(s)
- Maria‐Elena Torres‐Padilla
- Institute of Epigenetics and Stem Cells (IES)Helmholtz Zentrum MünchenMünchenGermany
- Faculty of BiologyLudwig‐Maximilians UniversitätMünchenGermany
| | - Annelien L Bredenoord
- Department of Medical HumanitiesJulius CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Karin R Jongsma
- Department of Medical HumanitiesJulius CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Astrid Lunkes
- Institute of Epigenetics and Stem Cells (IES)Helmholtz Zentrum MünchenMünchenGermany
- Institute of Functional EpigeneticsHelmholtz Zentrüm MünchenMünchenGermany
| | - Luca Marelli
- Life Sciences & Society LabCentre for Sociological ResearchKU LeuvenLeuvenBelgium
- Department of Experimental OncologyIEO, European Institute of OncologyIRCCSMilanItaly
- Department of Medical Biotechnologies and Translational MedicineUniversity of MilanMilanItaly
| | - Ines Pinheiro
- Nuclear Dynamics UnitInstitut CuriePSL Research UniversityParisFrance
| | - Giuseppe Testa
- Department of Experimental OncologyIEO, European Institute of OncologyIRCCSMilanItaly
- Department of Oncology and Hemato‐oncologyUniversità degli Studi di MilanoMilanItaly
- Human TechnopoleMilanItaly
| |
Collapse
|