301 results on '"J. Saez-Rodriguez"'
Search Results
2. P-319 Opposite functional alterations between aged endometria and that of women with uterine disorders offer plausible explanations to the increased incidence of uterine disorders with age
- Author
-
A Devesa Peiro, P Sebastian-Leon, F Garcia-Garcia, A Pellicer, J Saez-Rodriguez, and P Diaz-Gimeno
- Subjects
Reproductive Medicine ,Rehabilitation ,Obstetrics and Gynecology - Abstract
Study question Which is the functional relationship between age and uterine disorders at endometrial level? Summary answer Ciliogenesis and cell cycle processes were oppositely altered between the endometrium of patients with uterine disorders and that of > 35 y.o. women. What is known already Uterine disorders are complex and multifactorial conditions which incidence increases with age affecting women's reproductive health and fertility. Uterine disorders and age have been transcriptomically researched to identify potential biomarkers and underlying mechanisms in independent studies. However, there is a lack of studies comparing the effects caused by uterine disorders and age in endometrium to understand the functional relationship between them. The objective of this research was to compare the mechanisms underlying uterine disorders and age in the endometrium to understand the molecular relationship behind the increased incidence of these disorders with age. Study design, size, duration In silico study performed between 2016-2021 involving a systematic review at Gene Expression Omnibus sample repository to identify datasets with endometrial gene expression raw data associated to uterine disorders and age for answering the research question. Samples were preprocessed and analyzed with the same transcriptomic procedures for comparable analysis of functions affecting gene expression. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, DoRothEA and OmniPath functional databases were consulted for identifying functions/transcription factors involved. Participants/materials, setting, methods Nine endometrial transcriptomic datasets evaluating uterine disorders (123 cases, 127 controls) and one including control women with different ages (23–49 y.o., n = 27) were downloaded. Differentially expressed genes and gene set enrichment functional results related to uterine disorders or age for each dataset were calculated and integrated between uterine disorders under a meta-analysis with a random effects model to account for study heterogeneity. Upstream transcription factors and pathways were identified with footprinting methods. Main results and the role of chance All evaluated uterine disorders (adenocarcinoma (ADC), recurrent implantation failure (RIF), recurrent pregnancy loss (RPL) and eutopic endometriosis) shared a significant downregulation of six ciliary functions (FDR35 y.o. presented an opposite functional profile in comparison with the effect of uterine disorders - with a significant upregulation of 22 ciliary functions (FDR Limitations, reasons for caution This study depends on publicly available datasets to analyze. Although we considered potential confounding variables (time of biopsy collection, presence of benign pathologies in aged women) and study heterogeneity (using random effects models accounting for study variability), further studies are needed to corroborate our findings and test the proposed hypothesis. Wider implications of the findings A new hypothesis was generated regarding the molecular mechanisms behind the increased incidence of uterine disorders with age: With aging, the endometrium exhibits cell cycle arrest, inhibiting ciliogenesis. Consequently, compensatory mechanisms are activated to counteract aging-related alterations, but these mechanisms could be unbalanced towards the other extreme, originating distinct disorders. Trial registration number Not applicable
- Published
- 2022
- Full Text
- View/download PDF
3. S-25-02 Gene co-expression network analysis of toxicogenomic data linked to histopathology provides quantitative mode-of-action assessment and prediction of drug-induced organ toxicity
- Author
-
S.J. Kunnen, G. Callegaro, L.S. Wijaya, P. Trairatphisan, J. Mollon, S. Grosdidier, Y.W. Webster, J. Saez-Rodriguez, J.L. Stevens, and B. van de Water
- Subjects
General Medicine ,Toxicology - Published
- 2022
- Full Text
- View/download PDF
4. Logic modelling of toxicology pathways
- Author
-
J. Saez-Rodriguez and A Gabor
- Subjects
Engineering ,business.industry ,General Medicine ,Computational biology ,Toxicology ,business - Published
- 2021
- Full Text
- View/download PDF
5. S887 DISSECTING THE ROLE OF CXCL4 IN PRIMARY MYELOFIBROSIS
- Author
-
J. Saez-Rodriguez, H. Gleitz, S. Fuchs, A. Dugourd, J.E. Pritchard, Eric M.J. Bindels, Rebekka K. Schneider, R. Hoogenboezem, I. Snoeren, R. Kramann, B. Banjanin, and N. Leimkühler
- Subjects
Oncology ,medicine.medical_specialty ,Primary (chemistry) ,business.industry ,Internal medicine ,Medicine ,Hematology ,business ,Myelofibrosis ,medicine.disease - Published
- 2019
- Full Text
- View/download PDF
6. Characterizing and targeting glioblastoma neuron-tumor networks with retrograde tracing.
- Author
-
Tetzlaff SK, Reyhan E, Layer N, Bengtson CP, Heuer A, Schroers J, Faymonville AJ, Langeroudi AP, Drewa N, Keifert E, Wagner J, Soyka SJ, Schubert MC, Sivapalan N, Pramatarov RL, Buchert V, Wageringel T, Grabis E, Wißmann N, Alhalabi OT, Botz M, Bojcevski J, Campos J, Boztepe B, Scheck JG, Conic SH, Puschhof MC, Villa G, Drexler R, Zghaibeh Y, Hausmann F, Hänzelmann S, Karreman MA, Kurz FT, Schröter M, Thier M, Suwala AK, Forsberg-Nilsson K, Acuna C, Saez-Rodriguez J, Abdollahi A, Sahm F, Breckwoldt MO, Suchorska B, Ricklefs FL, Heiland DH, and Venkataramani V
- Abstract
Glioblastomas are invasive brain tumors with high therapeutic resistance. Neuron-to-glioma synapses have been shown to promote glioblastoma progression. However, a characterization of tumor-connected neurons has been hampered by a lack of technologies. Here, we adapted retrograde tracing using rabies viruses to investigate and manipulate neuron-tumor networks. Glioblastoma rapidly integrated into neural circuits across the brain, engaging in widespread functional communication, with cholinergic neurons driving glioblastoma invasion. We uncovered patient-specific and tumor-cell-state-dependent differences in synaptogenic gene expression associated with neuron-tumor connectivity and subsequent invasiveness. Importantly, radiotherapy enhanced neuron-tumor connectivity by increased neuronal activity. In turn, simultaneous neuronal activity inhibition and radiotherapy showed increased therapeutic effects, indicative of a role for neuron-to-glioma synapses in contributing to therapeutic resistance. Lastly, rabies-mediated genetic ablation of tumor-connected neurons halted glioblastoma progression, offering a viral strategy to tackle glioblastoma. Together, this study provides a framework to comprehensively characterize neuron-tumor networks and target glioblastoma., Competing Interests: Declaration of interests J.S.-R. reports funding from GSK, Pfizer, and Sanofi and fees/honoraria from Travere Therapeutics, Stadapharm, Astex, Owkin, Pfizer, and Grunenthal., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Single-cell transcriptomics reveal distinctive patterns of fibroblast activation in heart failure with preserved ejection fraction.
- Author
-
Lanzer JD, Wienecke LM, Ramirez Flores RO, Zylla MM, Kley C, Hartmann N, Sicklinger F, Schultz JH, Frey N, Saez-Rodriguez J, and Leuschner F
- Subjects
- Animals, Humans, Male, Mice, Fibrosis, Ventricular Function, Left, Female, Gene Expression Profiling, Heart Failure metabolism, Heart Failure physiopathology, Heart Failure genetics, Heart Failure pathology, Fibroblasts metabolism, Fibroblasts pathology, Stroke Volume, Transcriptome, Single-Cell Analysis, Disease Models, Animal, Mice, Inbred C57BL
- Abstract
Inflammation, fibrosis and metabolic stress critically promote heart failure with preserved ejection fraction (HFpEF). Exposure to high-fat diet and nitric oxide synthase inhibitor N[w]-nitro-l-arginine methyl ester (L-NAME) recapitulate features of HFpEF in mice. To identify disease-specific traits during adverse remodeling, we profiled interstitial cells in early murine HFpEF using single-cell RNAseq (scRNAseq). Diastolic dysfunction and perivascular fibrosis were accompanied by an activation of cardiac fibroblast and macrophage subsets. Integration of fibroblasts from HFpEF with two murine models for heart failure with reduced ejection fraction (HFrEF) identified a catalog of conserved fibroblast phenotypes across mouse models. Moreover, HFpEF-specific characteristics included induced metabolic, hypoxic and inflammatory transcription factors and pathways, including enhanced expression of Angiopoietin-like 4 (Angptl4) next to basement membrane compounds, such as collagen IV (Col4a1). Fibroblast activation was further dissected into transcriptional and compositional shifts and thereby highly responsive cell states for each HF model were identified. In contrast to HFrEF, where myofibroblast and matrifibrocyte activation were crucial features, we found that these cell states played a subsidiary role in early HFpEF. These disease-specific fibroblast signatures were corroborated in human myocardial bulk transcriptomes. Furthermore, we identified a potential cross-talk between macrophages and fibroblasts via SPP1 and TNFɑ with estimated fibroblast target genes including Col4a1 and Angptl4. Treatment with recombinant ANGPTL4 ameliorated the murine HFpEF phenotype and diastolic dysfunction by reducing collagen IV deposition from fibroblasts in vivo and in vitro. In line, ANGPTL4, was elevated in plasma samples of HFpEF patients and particularly high levels associated with a preserved global-longitudinal strain. Taken together, our study provides a comprehensive characterization of molecular fibroblast activation patterns in murine HFpEF, as well as the identification of Angiopoietin-like 4 as central mechanistic regulator with protective effects., Competing Interests: Declarations. Conflict of interest: JSR reports funding from GSK and Sanofi and fees from Travere Therapeutics, and Astex., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
8. Cell type mapping reveals tissue niches and interactions in subcortical multiple sclerosis lesions.
- Author
-
Lerma-Martin C, Badia-I-Mompel P, Ramirez Flores RO, Sekol P, Schäfer PSL, Riedl CJ, Hofmann A, Thäwel T, Wünnemann F, Ibarra-Arellano MA, Trobisch T, Eisele P, Schapiro D, Haeussler M, Hametner S, Saez-Rodriguez J, and Schirmer L
- Subjects
- Humans, Cell Communication physiology, Neuroglia pathology, Female, Male, Astrocytes pathology, Adult, Middle Aged, Brain pathology, Endothelial Cells pathology, Transcriptome, Multiple Sclerosis pathology
- Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system. Inflammation is gradually compartmentalized and restricted to specific tissue niches such as the lesion rim. However, the precise cell type composition of such niches, their interactions and changes between chronic active and inactive stages are incompletely understood. We used single-nucleus and spatial transcriptomics from subcortical MS and corresponding control tissues to map cell types and associated pathways to lesion and nonlesion areas. We identified niches such as perivascular spaces, the inflamed lesion rim or the lesion core that are associated with the glial scar and a cilia-forming astrocyte subtype. Focusing on the inflamed rim of chronic active lesions, we uncovered cell-cell communication events between myeloid, endothelial and glial cell types. Our results provide insight into the cellular composition, multicellular programs and intercellular communication in tissue niches along the conversion from a homeostatic to a dysfunctional state underlying lesion progression in MS., Competing Interests: Competing interests: P.E. has received travel expenses from Bayer Health Care. D.S. reports funding from Cellzome, a GSK company and received honorariums from Immunai, Noetik, Alpenglow and Lunaphore. J.S.-R. reports funding from GSK, Pfizer and Sanofi and fees/honoraria from Travere Therapeutics, Stadapharm, Astex, Owkin, Moderna, Pfizer and Grunenthal. L.S. reports research support from Roche and Merck and filed a patent for the detection of antibodies against KIR4.1 in a subpopulation of patients with multiple sclerosis (WO2015166057A1). The remaining authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. PhosX: data-driven kinase activity inference from phosphoproteomics experiments.
- Author
-
Lussana A, Müller-Dott S, Saez-Rodriguez J, and Petsalaki E
- Subjects
- Humans, Phosphoproteins metabolism, Phosphorylation, Substrate Specificity, Phosphopeptides metabolism, Phosphopeptides analysis, Protein Kinases metabolism, Computational Biology methods, Algorithms, Proteomics methods, Software
- Abstract
Summary: The inference of kinase activity from phosphoproteomics data can point to causal mechanisms driving signalling processes and potential drug targets. Identifying the kinases whose change in activity explains the observed phosphorylation profiles, however, remains challenging, and constrained by the manually curated knowledge of kinase-substrate associations. Recently, experimentally determined substrate sequence specificities of human kinases have become available, but robust methods to exploit this new data for kinase activity inference are still missing. We present PhosX, a method to estimate differential kinase activity from phosphoproteomics data that combines state-of-the-art statistics in enrichment analysis with kinases' substrate sequence specificity information. Using a large phosphoproteomics dataset with known differentially regulated kinases we show that our method identifies upregulated and downregulated kinases by only relying on the input phosphopeptides' sequences and intensity changes. We find that PhosX outperforms the currently available approach for the same task, and performs better or similarly to state-of-the-art methods that rely on previously known kinase-substrate associations. We therefore recommend its use for data-driven kinase activity inference., Availability and Implementation: PhosX is implemented in Python, open-source under the Apache-2.0 licence, and distributed on the Python Package Index. The code is available on GitHub (https://github.com/alussana/phosx)., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
10. Author Correction: Community assessment of methods to deconvolve cellular composition from bulk gene expression.
- Author
-
White BS, de Reyniès A, Newman AM, Waterfall JJ, Lamb A, Petitprez F, Lin Y, Yu R, Guerrero-Gimenez ME, Domanskyi S, Monaco G, Chung V, Banerjee J, Derrick D, Valdeolivas A, Li H, Xiao X, Wang S, Zheng F, Yang W, Catania CA, Lang BJ, Bertus TJ, Piermarocchi C, Caruso FP, Ceccarelli M, Yu T, Guo X, Bletz J, Coller J, Maecker H, Duault C, Shokoohi V, Patel S, Liliental JE, Simon S, Saez-Rodriguez J, Heiser LM, Guinney J, and Gentles AJ
- Published
- 2024
- Full Text
- View/download PDF
11. Single-cell integration reveals metaplasia in inflammatory gut diseases.
- Author
-
Oliver AJ, Huang N, Bartolome-Casado R, Li R, Koplev S, Nilsen HR, Moy M, Cakir B, Polanski K, Gudiño V, Melón-Ardanaz E, Sumanaweera D, Dimitrov D, Milchsack LM, FitzPatrick MEB, Provine NM, Boccacino JM, Dann E, Predeus AV, To K, Prete M, Chapman JA, Masi AC, Stephenson E, Engelbert J, Lobentanzer S, Perera S, Richardson L, Kapuge R, Wilbrey-Clark A, Semprich CI, Ellams S, Tudor C, Joseph P, Garrido-Trigo A, Corraliza AM, Oliver TRW, Hook CE, James KR, Mahbubani KT, Saeb-Parsy K, Zilbauer M, Saez-Rodriguez J, Høivik ML, Bækkevold ES, Stewart CJ, Berrington JE, Meyer KB, Klenerman P, Salas A, Haniffa M, Jahnsen FL, Elmentaite R, and Teichmann SA
- Subjects
- Adult, Humans, Brunner Glands metabolism, Case-Control Studies, Celiac Disease pathology, Colitis, Ulcerative pathology, Crohn Disease pathology, Crohn Disease immunology, Datasets as Topic, Gastric Mucosa immunology, Gastric Mucosa pathology, Gastrointestinal Neoplasms immunology, Gastrointestinal Neoplasms pathology, Gastrointestinal Tract immunology, Gastrointestinal Tract pathology, Health, Intestinal Mucosa immunology, Intestinal Mucosa pathology, Neutrophils immunology, Pylorus metabolism, Quality Control, Single-Cell Gene Expression Analysis, Stem Cells immunology, Stem Cells metabolism, Stem Cells pathology, T-Lymphocytes immunology, Child, Epithelial Cells pathology, Gastrointestinal Diseases pathology, Gastrointestinal Diseases immunology, Inflammation pathology, Inflammation immunology, Metaplasia pathology, Single-Cell Analysis methods
- Abstract
The gastrointestinal tract is a multi-organ system crucial for efficient nutrient uptake and barrier immunity. Advances in genomics and a surge in gastrointestinal diseases
1,2 has fuelled efforts to catalogue cells constituting gastrointestinal tissues in health and disease3 . Here we present systematic integration of 25 single-cell RNA sequencing datasets spanning the entire healthy gastrointestinal tract in development and in adulthood. We uniformly processed 385 samples from 189 healthy controls using a newly developed automated quality control approach (scAutoQC), leading to a healthy reference atlas with approximately 1.1 million cells and 136 fine-grained cell states. We anchor 12 gastrointestinal disease datasets spanning gastrointestinal cancers, coeliac disease, ulcerative colitis and Crohn's disease to this reference. Utilizing this 1.6 million cell resource (gutcellatlas.org), we discover epithelial cell metaplasia originating from stem cells in intestinal inflammatory diseases with transcriptional similarity to cells found in pyloric and Brunner's glands. Although previously linked to mucosal healing4 , we now implicate pyloric gland metaplastic cells in inflammation through recruitment of immune cells including T cells and neutrophils. Overall, we describe inflammation-induced changes in stem cells that alter mucosal tissue architecture and promote further inflammation, a concept applicable to other tissues and diseases., Competing Interests: Competing interests: S.A.T. is a scientific advisory board member of ForeSite Labs, OMass Therapeutics, a co-founder and equity holder of TransitionBio and EnsoCell Therapeutics, a non-executive director of 10x Genomics and a part-time employee of GlaxoSmithKline. R.E. is an equity holder in EnsoCell. P.K. has consulted for AstraZeneca, UCB, Biomunex and Infinitopes. N.M.P reports consulting fees from Infinitopes. J.S.-R. reports funding from GSK, Pfizer and Sanofi and fees/honoraria from Travere Therapeutics, Stadapharm, Astex, Owkin, Pfizer, Moderna and Grunenthal. A.S. is the recipient of research grants from Roche-Genentech, Abbvie, GSK, Scipher Medicine, Pfizer, Alimentiv, Boehringer Ingelheim and Agomab and has received consulting fees from Genentech, GSK, Pfizer, HotSpot Therapeutics, Alimentiv, Agomab, Goodgut and Orikine. R.E. and S.A.T are inventors on the patent GB2412853.0 filed in the UK, some components of which are related to this work. All other authors declare no competing interests., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
12. Defining the molecular response to ischemia-reperfusion injury and remote ischemic preconditioning in human kidney transplantation.
- Author
-
Nordström J, Badia-I-Mompel P, Witasp A, Schwarz A, Evenepoel P, Moor MB, Wennberg L, Saez-Rodriguez J, Wernerson A, and Olauson H
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Living Donors, Kidney metabolism, Transcriptome, Kynurenine blood, Kynurenine metabolism, Kidney Transplantation adverse effects, Reperfusion Injury prevention & control, Reperfusion Injury metabolism, Ischemic Preconditioning methods
- Abstract
Background: Ischemia-reperfusion injury (IRI) inevitably occurs during kidney transplantation and extended ischemia is associated with delayed graft function and poor outcomes. Remote ischemic preconditioning (RIPC) is a simple, noninvasive procedure aimed at reducing IRI and improving graft function. Experimental studies have implicated the kynurenine pathway as a protective mechanism behind RIPC., Methods: First, paired biopsies from 11 living kidney donors were analyzed to characterize the acute transcriptomic response to IRI. Second, 16 living kidney donors were subjected to either RIPC (n = 9) or no pretreatment (n = 7) to evaluate the impact of RIPC on the transcriptomic response to IRI. Finally, the effect of RIPC on plasma metabolites was analyzed in 49 healthy subjects., Results: There was a robust immediate response to IRI in the renal transcriptomes of living-donor kidney transplantation, including activation of the mitogen-activated protein kinase (MAPK) and epidermal growth factor receptor (EGFR) pathways. Preconditioning with RIPC did not significantly alter the transcriptomic response to IRI or the concentration of plasma metabolites., Conclusions: The present data validate living-donor kidney transplantation as a suitable model for mechanistic studies of IRI in human kidneys. The failure of RIPC to alter transcriptomic responses or metabolites in the kynurenine pathway raises the question of the robustness of the standard procedure used to induce RIPC, and might explain the mixed results in clinical trials evaluating RIPC as a method to attenuate IRI., Competing Interests: J.S.R. reports funding from GSK and Sanofi and fees from Travere Therapeutics and Astex. This does not alter our adherence to PLOS ONE policies on sharing data and materials. There are no patents, products in development, or marketed products associated with this research to declare., (Copyright: © 2024 Nordström et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
13. Author Correction: Compartments in medulloblastoma with extensive nodularity are connected through differentiation along the granular precursor lineage.
- Author
-
Ghasemi DR, Okonechnikov K, Rademacher A, Tirier S, Maass KK, Schumacher H, Joshi P, Gold MP, Sundheimer J, Statz B, Rifaioglu AS, Bauer K, Schumacher S, Bortolomeazzi M, Giangaspero F, Ernst KJ, Clifford SC, Saez-Rodriguez J, Jones DTW, Kawauchi D, Fraenkel E, Mallm JP, Rippe K, Korshunov A, Pfister SM, and Pajtler KW
- Published
- 2024
- Full Text
- View/download PDF
14. LIANA+ provides an all-in-one framework for cell-cell communication inference.
- Author
-
Dimitrov D, Schäfer PSL, Farr E, Rodriguez-Mier P, Lobentanzer S, Badia-I-Mompel P, Dugourd A, Tanevski J, Ramirez Flores RO, and Saez-Rodriguez J
- Subjects
- Humans, Software, Animals, Transcriptome, Computational Biology methods, Gene Expression Profiling methods, Cell Communication, Single-Cell Analysis methods, Signal Transduction
- Abstract
The growing availability of single-cell and spatially resolved transcriptomics has led to the development of many approaches to infer cell-cell communication, each capturing only a partial view of the complex landscape of intercellular signalling. Here we present LIANA+, a scalable framework built around a rich knowledge base to decode coordinated inter- and intracellular signalling events from single- and multi-condition datasets in both single-cell and spatially resolved data. By extending and unifying established methodologies, LIANA+ provides a comprehensive set of synergistic components to study cell-cell communication via diverse molecular mediators, including those measured in multi-omics data. LIANA+ is accessible at https://github.com/saezlab/liana-py with extensive vignettes ( https://liana-py.readthedocs.io/ ) and provides an all-in-one solution to intercellular communication inference., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
15. Gene regulatory networks in disease and ageing.
- Author
-
Unger Avila P, Padvitski T, Leote AC, Chen H, Saez-Rodriguez J, Kann M, and Beyer A
- Subjects
- Humans, Gene Expression Regulation, Gene Regulatory Networks, Aging genetics
- Abstract
The precise control of gene expression is required for the maintenance of cellular homeostasis and proper cellular function, and the declining control of gene expression with age is considered a major contributor to age-associated changes in cellular physiology and disease. The coordination of gene expression can be represented through models of the molecular interactions that govern gene expression levels, so-called gene regulatory networks. Gene regulatory networks can represent interactions that occur through signal transduction, those that involve regulatory transcription factors, or statistical models of gene-gene relationships based on the premise that certain sets of genes tend to be coexpressed across a range of conditions and cell types. Advances in experimental and computational technologies have enabled the inference of these networks on an unprecedented scale and at unprecedented precision. Here, we delineate different types of gene regulatory networks and their cell-biological interpretation. We describe methods for inferring such networks from large-scale, multi-omics datasets and present applications that have aided our understanding of cellular ageing and disease mechanisms., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
16. Community assessment of methods to deconvolve cellular composition from bulk gene expression.
- Author
-
White BS, de Reyniès A, Newman AM, Waterfall JJ, Lamb A, Petitprez F, Lin Y, Yu R, Guerrero-Gimenez ME, Domanskyi S, Monaco G, Chung V, Banerjee J, Derrick D, Valdeolivas A, Li H, Xiao X, Wang S, Zheng F, Yang W, Catania CA, Lang BJ, Bertus TJ, Piermarocchi C, Caruso FP, Ceccarelli M, Yu T, Guo X, Bletz J, Coller J, Maecker H, Duault C, Shokoohi V, Patel S, Liliental JE, Simon S, Saez-Rodriguez J, Heiser LM, Guinney J, and Gentles AJ
- Subjects
- Humans, Gene Expression Profiling methods, Transcriptome, Deep Learning, Computational Biology methods, Lymphocytes, Tumor-Infiltrating immunology, Gene Expression Regulation, Neoplastic, CD8-Positive T-Lymphocytes metabolism, CD4-Positive T-Lymphocytes metabolism, Neoplasms genetics, Neoplasms immunology, Neoplasms pathology
- Abstract
We evaluate deconvolution methods, which infer levels of immune infiltration from bulk expression of tumor samples, through a community-wide DREAM Challenge. We assess six published and 22 community-contributed methods using in vitro and in silico transcriptional profiles of admixed cancer and healthy immune cells. Several published methods predict most cell types well, though they either were not trained to evaluate all functional CD8+ T cell states or do so with low accuracy. Several community-contributed methods address this gap, including a deep learning-based approach, whose strong performance establishes the applicability of this paradigm to deconvolution. Despite being developed largely using immune cells from healthy tissues, deconvolution methods predict levels of tumor-derived immune cells well. Our admixed and purified transcriptional profiles will be a valuable resource for developing deconvolution methods, including in response to common challenges we observe across methods, such as sensitive identification of functional CD4+ T cell states., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
17. Endogenous adenine is a potential driver of the cardiovascular-kidney-metabolic syndrome.
- Author
-
Tamayo I, Lee HJ, Aslam MI, Liu JJ, Ragi N, Karanam V, Maity S, Saliba A, Treviño E, Zheng H, Lim SC, Lanzer JD, Bjornstad P, Tuttle K, Bedi KC Jr, Margulies KB, Ramachandran V, Abdel-Latif A, Saez-Rodriguez J, Iyengar R, Bopassa JC, and Sharma K
- Abstract
Mechanisms underlying the cardiovascular-kidney-metabolic (CKM) syndrome are unknown, although key small molecule metabolites may be involved. Bulk and spatial metabolomics identified adenine to be upregulated and specifically enriched in coronary blood vessels in hearts from patients with diabetes and left ventricular hypertrophy. Single nucleus gene expression studies revealed that endothelial methylthioadenosine phosphorylase (MTAP) was increased in human hearts with hypertrophic cardiomyopathy. The urine adenine/creatinine ratio in patients was predictive of incident heart failure with preserved ejection fraction. Heart adenine and MTAP gene expression was increased in a 2-hit mouse model of hypertrophic heart disease and in a model of diastolic dysfunction with diabetes. Inhibition of MTAP blocked adenine accumulation in the heart, restored heart dysfunction in mice with type 2 diabetes and prevented ischemic heart damage in a rat model of myocardial infarction. Mechanistically, adenine-induced impaired mitophagy was reversed by reduction of mTOR. These studies indicate that endogenous adenine is in a causal pathway for heart failure and ischemic heart disease in the context of CKM syndrome., Competing Interests: Competing interests: Dr. Margulies holds research grants from Amgen and serves as a scientific consultant/advisory board member for Bristol Myers Squibb and Amgen. Dr. Sharma serves on the data safety board for Cara Therapeutics and holds equity in SygnaMap. All other authors declare that they have no competing interests. Dr. Tuttle has received investigator-initiated grant support (to Providence Inland Northwest Health) from Travere and Bayer outside of the submitted work; consultancy fees from AstraZeneca, Boehringer Ingelheim, Eli Lilly and Company, Novo Nordisk and Travere; speaker fees from AstraZeneca, Eli Lilly, and Novo Nordisk. Dr. Julia Saez-Rodriguez reports funding from GSK, Pfizer and Sanofi & fees/honoraria from Travere Therapeutics, Stadapharm, Astex, Owkin, Pfizer and Grunenthal.
- Published
- 2024
- Full Text
- View/download PDF
18. Assessing the impact of transcriptomics data analysis pipelines on downstream functional enrichment results.
- Author
-
Paton V, Ramirez Flores RO, Gabor A, Badia-I-Mompel P, Tanevski J, Garrido-Rodriguez M, and Saez-Rodriguez J
- Subjects
- Humans, Cell Line, Tumor, Software, Heart Failure genetics, Workflow, Neoplasms genetics, Data Analysis, Benchmarking, Gene Expression Profiling methods, Transcriptome genetics
- Abstract
Transcriptomics is widely used to assess the state of biological systems. There are many tools for the different steps, such as normalization, differential expression, and enrichment. While numerous studies have examined the impact of method choices on differential expression results, little attention has been paid to their effects on further downstream functional analysis, which typically provides the basis for interpretation and follow-up experiments. To address this, we introduce FLOP, a comprehensive nextflow-based workflow combining methods to perform end-to-end analyses of transcriptomics data. We illustrate FLOP on datasets ranging from end-stage heart failure patients to cancer cell lines. We discovered effects not noticeable at the gene-level, and observed that not filtering the data had the highest impact on the correlation between pipelines in the gene set space. Moreover, we performed three benchmarks to evaluate the 12 pipelines included in FLOP, and confirmed that filtering is essential in scenarios of expected moderate-to-low biological signal. Overall, our results underscore the impact of carefully evaluating the consequences of the choice of preprocessing methods on downstream enrichment analyses. We envision FLOP as a valuable tool to measure the robustness of functional analyses, ultimately leading to more reliable and conclusive biological findings., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
19. Molecular causality in the advent of foundation models.
- Author
-
Lobentanzer S, Rodriguez-Mier P, Bauer S, and Saez-Rodriguez J
- Subjects
- Humans, Biomedical Research, Models, Biological, Systems Biology, Machine Learning, Causality
- Abstract
Correlation is not causation: this simple and uncontroversial statement has far-reaching implications. Defining and applying causality in biomedical research has posed significant challenges to the scientific community. In this perspective, we attempt to connect the partly disparate fields of systems biology, causal reasoning, and machine learning to inform future approaches in the field of systems biology and molecular medicine., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
20. A single-sample workflow for joint metabolomic and proteomic analysis of clinical specimens.
- Author
-
Gegner HM, Naake T, Aljakouch K, Dugourd A, Kliewer G, Müller T, Schilling D, Schneider MA, Kunze-Rohrbach N, Grünewald TGP, Hell R, Saez-Rodriguez J, Huber W, Poschet G, and Krijgsveld J
- Abstract
Understanding the interplay of the proteome and the metabolome helps to understand cellular regulation and response. To enable robust inferences from such multi-omics analyses, we introduced and evaluated a workflow for combined proteome and metabolome analysis starting from a single sample. Specifically, we integrated established and individually optimized protocols for metabolomic and proteomic profiling (EtOH/MTBE and autoSP3, respectively) into a unified workflow (termed MTBE-SP3), and took advantage of the fact that the protein residue of the metabolomic sample can be used as a direct input for proteome analysis. We particularly evaluated the performance of proteome analysis in MTBE-SP3, and demonstrated equivalence of proteome profiles irrespective of prior metabolite extraction. In addition, MTBE-SP3 combines the advantages of EtOH/MTBE and autoSP3 for semi-automated metabolite extraction and fully automated proteome sample preparation, respectively, thus advancing standardization and scalability for large-scale studies. We showed that MTBE-SP3 can be applied to various biological matrices (FFPE tissue, fresh-frozen tissue, plasma, serum and cells) to enable implementation in a variety of clinical settings. To demonstrate applicability, we applied MTBE-SP3 and autoSP3 to a lung adenocarcinoma cohort showing consistent proteomic alterations between tumour and non-tumour adjacent tissue independent of the method used. Integration with metabolomic data obtained from the same samples revealed mitochondrial dysfunction in tumour tissue through deregulation of OGDH, SDH family enzymes and PKM. In summary, MTBE-SP3 enables the facile and reliable parallel measurement of proteins and metabolites obtained from the same sample, benefiting from reduced sample variation and input amount. This workflow is particularly applicable for studies with limited sample availability and offers the potential to enhance the integration of metabolomic and proteomic datasets., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
21. Drugst.One - a plug-and-play solution for online systems medicine and network-based drug repurposing.
- Author
-
Maier A, Hartung M, Abovsky M, Adamowicz K, Bader GD, Baier S, Blumenthal DB, Chen J, Elkjaer ML, Garcia-Hernandez C, Helmy M, Hoffmann M, Jurisica I, Kotlyar M, Lazareva O, Levi H, List M, Lobentanzer S, Loscalzo J, Malod-Dognin N, Manz Q, Matschinske J, Mee M, Oubounyt M, Pastrello C, Pico AR, Pillich RT, Poschenrieder JM, Pratt D, Pržulj N, Sadegh S, Saez-Rodriguez J, Sarkar S, Shaked G, Shamir R, Trummer N, Turhan U, Wang RS, Zolotareva O, and Baumbach J
- Subjects
- Humans, Internet, Drug Discovery methods, Systems Biology methods, Computational Biology methods, Drug Repositioning methods, Software
- Abstract
In recent decades, the development of new drugs has become increasingly expensive and inefficient, and the molecular mechanisms of most pharmaceuticals remain poorly understood. In response, computational systems and network medicine tools have emerged to identify potential drug repurposing candidates. However, these tools often require complex installation and lack intuitive visual network mining capabilities. To tackle these challenges, we introduce Drugst.One, a platform that assists specialized computational medicine tools in becoming user-friendly, web-based utilities for drug repurposing. With just three lines of code, Drugst.One turns any systems biology software into an interactive web tool for modeling and analyzing complex protein-drug-disease networks. Demonstrating its broad adaptability, Drugst.One has been successfully integrated with 21 computational systems medicine tools. Available at https://drugst.one, Drugst.One has significant potential for streamlining the drug discovery process, allowing researchers to focus on essential aspects of pharmaceutical treatment research., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
22. Opening the Black Box: Spatial Transcriptomics and the Relevance of Artificial Intelligence-Detected Prognostic Regions in High-Grade Serous Carcinoma.
- Author
-
Laury AR, Zheng S, Aho N, Fallegger R, Hänninen S, Saez-Rodriguez J, Tanevski J, Youssef O, Tang J, and Carpén OM
- Subjects
- Humans, Female, Prognosis, Gene Expression Profiling methods, Middle Aged, Aged, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Ovarian Neoplasms drug therapy, Transcriptome, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous pathology, Cystadenocarcinoma, Serous drug therapy, Artificial Intelligence
- Abstract
Image-based deep learning models are used to extract new information from standard hematoxylin and eosin pathology slides; however, biological interpretation of the features detected by artificial intelligence (AI) remains a challenge. High-grade serous carcinoma of the ovary (HGSC) is characterized by aggressive behavior and chemotherapy resistance, but also exhibits striking variability in outcome. Our understanding of this disease is limited, partly due to considerable tumor heterogeneity. We previously trained an AI model to identify HGSC tumor regions that are highly associated with outcome status but are indistinguishable by conventional morphologic methods. Here, we applied spatially resolved transcriptomics to further profile the AI-identified tumor regions in 16 patients (8 per outcome group) and identify molecular features related to disease outcome in patients who underwent primary debulking surgery and platinum-based chemotherapy. We examined formalin-fixed paraffin-embedded tissue from (1) regions identified by the AI model as highly associated with short or extended chemotherapy response, and (2) background tumor regions (not identified by the AI model as highly associated with outcome status) from the same tumors. We show that the transcriptomic profiles of AI-identified regions are more distinct than background regions from the same tumors, are superior in predicting outcome, and differ in several pathways including those associated with chemoresistance in HGSC. Further, we find that poor outcome and good outcome regions are enriched by different tumor subpopulations, suggesting distinctive interaction patterns. In summary, our work presents proof of concept that AI-guided spatial transcriptomic analysis improves recognition of biologic features relevant to patient outcomes., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
23. DOT: a flexible multi-objective optimization framework for transferring features across single-cell and spatial omics.
- Author
-
Rahimi A, Vale-Silva LA, Fälth Savitski M, Tanevski J, and Saez-Rodriguez J
- Subjects
- Humans, Gene Expression Profiling methods, Transcriptome, Animals, Computational Biology methods, Single-Cell Analysis methods, Algorithms
- Abstract
Single-cell transcriptomics and spatially-resolved imaging/sequencing technologies have revolutionized biomedical research. However, they suffer from lack of spatial information and a trade-off of resolution and gene coverage, respectively. We propose DOT, a multi-objective optimization framework for transferring cellular features across these data modalities, thus integrating their complementary information. DOT uses genes beyond those common to the data modalities, exploits the local spatial context, transfers spatial features beyond cell-type information, and infers absolute/relative abundance of cell populations at tissue locations. Thus, DOT bridges single-cell transcriptomics data with both high- and low-resolution spatially-resolved data. Moreover, DOT combines practical aspects related to cell composition, heterogeneity, technical effects, and integration of prior knowledge. Our fast implementation based on the Frank-Wolfe algorithm achieves state-of-the-art or improved performance in localizing cell features in high- and low-resolution spatial data and estimating the expression of unmeasured genes in low-coverage spatial data., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
24. Spatially resolved multiomics on the neuronal effects induced by spaceflight in mice.
- Author
-
Masarapu Y, Cekanaviciute E, Andrusivova Z, Westholm JO, Björklund Å, Fallegger R, Badia-I-Mompel P, Boyko V, Vasisht S, Saravia-Butler A, Gebre S, Lázár E, Graziano M, Frapard S, Hinshaw RG, Bergmann O, Taylor DM, Wallace DC, Sylvén C, Meletis K, Saez-Rodriguez J, Galazka JM, Costes SV, and Giacomello S
- Subjects
- Animals, Mice, Female, Transcriptome, Neurogenesis, Single-Cell Analysis, Mice, Inbred C57BL, Synaptic Transmission, Weightlessness adverse effects, Astrocytes metabolism, Oxidative Stress, Gene Expression Profiling, Multiomics, Space Flight, Brain metabolism, Brain pathology, Neurons metabolism
- Abstract
Impairment of the central nervous system (CNS) poses a significant health risk for astronauts during long-duration space missions. In this study, we employed an innovative approach by integrating single-cell multiomics (transcriptomics and chromatin accessibility) with spatial transcriptomics to elucidate the impact of spaceflight on the mouse brain in female mice. Our comparative analysis between ground control and spaceflight-exposed animals revealed significant alterations in essential brain processes including neurogenesis, synaptogenesis and synaptic transmission, particularly affecting the cortex, hippocampus, striatum and neuroendocrine structures. Additionally, we observed astrocyte activation and signs of immune dysfunction. At the pathway level, some spaceflight-induced changes in the brain exhibit similarities with neurodegenerative disorders, marked by oxidative stress and protein misfolding. Our integrated spatial multiomics approach serves as a stepping stone towards understanding spaceflight-induced CNS impairments at the level of individual brain regions and cell types, and provides a basis for comparison in future spaceflight studies. For broader scientific impact, all datasets from this study are available through an interactive data portal, as well as the National Aeronautics and Space Administration (NASA) Open Science Data Repository (OSDR)., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
25. MetalinksDB: a flexible and contextualizable resource of metabolite-protein interactions.
- Author
-
Farr E, Dimitrov D, Schmidt C, Turei D, Lobentanzer S, Dugourd A, and Saez-Rodriguez J
- Subjects
- Humans, Cell Communication, Kidney Neoplasms metabolism, Kidney Neoplasms genetics, Acute Kidney Injury metabolism, Acute Kidney Injury genetics, Computational Biology methods, Proteins metabolism, Proteins genetics, Software, Transcriptome, Signal Transduction
- Abstract
From the catalytic breakdown of nutrients to signaling, interactions between metabolites and proteins play an essential role in cellular function. An important case is cell-cell communication, where metabolites, secreted into the microenvironment, initiate signaling cascades by binding to intra- or extracellular receptors of neighboring cells. Protein-protein cell-cell communication interactions are routinely predicted from transcriptomic data. However, inferring metabolite-mediated intercellular signaling remains challenging, partially due to the limited size of intercellular prior knowledge resources focused on metabolites. Here, we leverage knowledge-graph infrastructure to integrate generalistic metabolite-protein with curated metabolite-receptor resources to create MetalinksDB. MetalinksDB is an order of magnitude larger than existing metabolite-receptor resources and can be tailored to specific biological contexts, such as diseases, pathways, or tissue/cellular locations. We demonstrate MetalinksDB's utility in identifying deregulated processes in renal cancer using multi-omics bulk data. Furthermore, we infer metabolite-driven intercellular signaling in acute kidney injury using spatial transcriptomics data. MetalinksDB is a comprehensive and customizable database of intercellular metabolite-protein interactions, accessible via a web interface (https://metalinks.omnipathdb.org/) and programmatically as a knowledge graph (https://github.com/biocypher/metalinks). We anticipate that by enabling diverse analyses tailored to specific biological contexts, MetalinksDB will facilitate the discovery of disease-relevant metabolite-mediated intercellular signaling processes., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
26. Complementing Cell Taxonomies with a Multicellular Analysis of Tissues.
- Author
-
Ramirez Flores RO, Schäfer PSL, Küchenhoff L, and Saez-Rodriguez J
- Subjects
- Cells, Tissues cytology
- Abstract
The application of single-cell molecular profiling coupled with spatial technologies has enabled charting of cellular heterogeneity in reference tissues and in disease. This new wave of molecular data has highlighted the expected diversity of single-cell dynamics upon shared external queues and spatial organizations. However, little is known about the relationship between single-cell heterogeneity and the emergence and maintenance of robust multicellular processes in developed tissues and its role in (patho)physiology. Here, we present emerging computational modeling strategies that use increasingly available large-scale cross-condition single-cell and spatial datasets to study multicellular organization in tissues and complement cell taxonomies. This perspective should enable us to better understand how cells within tissues collectively process information and adapt synchronized responses in disease contexts and to bridge the gap between structural changes and functions in tissues.
- Published
- 2024
- Full Text
- View/download PDF
27. Combining LIANA and Tensor-cell2cell to decipher cell-cell communication across multiple samples.
- Author
-
Baghdassarian HM, Dimitrov D, Armingol E, Saez-Rodriguez J, and Lewis NE
- Subjects
- Humans, Computational Biology methods, Single-Cell Analysis methods, Cell Communication physiology, Software
- Abstract
In recent years, data-driven inference of cell-cell communication has helped reveal coordinated biological processes across cell types. Here, we integrate two tools, LIANA and Tensor-cell2cell, which, when combined, can deploy multiple existing methods and resources to enable the robust and flexible identification of cell-cell communication programs across multiple samples. In this work, we show how the integration of our tools facilitates the choice of method to infer cell-cell communication and subsequently perform an unsupervised deconvolution to obtain and summarize biological insights. We explain how to perform the analysis step by step in both Python and R and provide online tutorials with detailed instructions available at https://ccc-protocols.readthedocs.io/. This workflow typically takes ∼1.5 h to complete from installation to downstream visualizations on a graphics processing unit-enabled computer for a dataset of ∼63,000 cells, 10 cell types, and 12 samples., Competing Interests: Declaration of interests J.S.-R. reports funding from GSK, Pfizer, and Sanofi and fees/honoraria from Travere Therapeutics, Stadapharm, Astex, Pfizer, and Grunenthal. N.E.L. reports funding during the course of this work from Sanofi, Amgen, Sartorius, and Ionis and is a co-founder of NeuImmune, Inc., and Augment Biologics., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
28. Defining and benchmarking open problems in single-cell analysis.
- Author
-
Luecken MD, Gigante S, Burkhardt DB, Cannoodt R, Strobl DC, Markov NS, Zappia L, Palla G, Lewis W, Dimitrov D, Vinyard ME, Magruder DS, Andersson A, Dann E, Qin Q, Otto DJ, Klein M, Botvinnik OB, Deconinck L, Waldrant K, Bloom JM, Pisco AO, Saez-Rodriguez J, Wulsin D, Pinello L, Saeys Y, Theis FJ, and Krishnaswamy S
- Abstract
With the growing number of single-cell analysis tools, benchmarks are increasingly important to guide analysis and method development. However, a lack of standardisation and extensibility in current benchmarks limits their usability, longevity, and relevance to the community. We present Open Problems, a living, extensible, community-guided benchmarking platform including 10 current single-cell tasks that we envision will raise standards for the selection, evaluation, and development of methods in single-cell analysis., Competing Interests: M.D.L. contracted for the Chan Zuckerberg Initiative and received speaker fees from Pfizer and Janssen Pharmaceuticals. S.G. has equity interest in Immunai Inc. D.B.B. is a paid employee of and has equity interest in Cellarity Inc. L.Z. has consulted for Lamin Labs GmbH. W.L. contracted for Protein Evolution Incorporated. From 2019 to 2022 A.A. was a consultant for 10X Genomics. From October 2023 E.D. has been a consultant for EnsoCell Therapeutics. O.B.B is currently an employee of Bridge Bio Pharma. J.B. has equity interest in Cellarity, Inc. J.S.R. reports funding from GSK, Pfizer and Sanofi and fees/honoraria from Travere Therapeutics, Stadapharm, Astex, Owkin, Pfizer and Grunenthal. D.W. has equity interest in Immunai Inc. F.J.T. consults for Immunai Inc., Singularity Bio B.V., CytoReason Ltd, Cellarity, and has ownership interest in Dermagnostix GmbH and Cellarity. S.K. is a visiting professor at Meta and scientific advisor at Ascent Bio, Inc. E.d.V.B has ownership interest in Retro Biosciences and ImYoo Inc and is employed by ImYoo Inc. A.T.C. is an employee of Orion Medicines. B.D. is a paid employee of and has equity interest in Cellarity Inc. A.G. is currently an employee of Google DeepMind. Google DeepMind has not directed any aspect of this study nor exerts any commercial rights over the results. R.L. is an employee of Genentech. V.S. has ownership interest in Altos Labs and Vesalius Therapeutics. A.T. has an ownership interest in Dreamfold.
- Published
- 2024
- Full Text
- View/download PDF
29. Network integration of thermal proteome profiling with multi-omics data decodes PARP inhibition.
- Author
-
Burtscher ML, Gade S, Garrido-Rodriguez M, Rutkowska A, Werner T, Eberl HC, Petretich M, Knopf N, Zirngibl K, Grandi P, Bergamini G, Bantscheff M, Fälth-Savitski M, and Saez-Rodriguez J
- Subjects
- Multiomics, Poly(ADP-ribose) Polymerases genetics, Poly(ADP-ribose) Polymerases metabolism, Proteomics methods, Proteome, Poly(ADP-ribose) Polymerase Inhibitors pharmacology
- Abstract
Complex disease phenotypes often span multiple molecular processes. Functional characterization of these processes can shed light on disease mechanisms and drug effects. Thermal Proteome Profiling (TPP) is a mass-spectrometry (MS) based technique assessing changes in thermal protein stability that can serve as proxies of functional protein changes. These unique insights of TPP can complement those obtained by other omics technologies. Here, we show how TPP can be integrated with phosphoproteomics and transcriptomics in a network-based approach using COSMOS, a multi-omics integration framework, to provide an integrated view of transcription factors, kinases and proteins with altered thermal stability. This allowed us to recover consequences of Poly (ADP-ribose) polymerase (PARP) inhibition in ovarian cancer cells on cell cycle and DNA damage response as well as interferon and hippo signaling. We found that TPP offers a complementary perspective to other omics data modalities, and that its integration allowed us to obtain a more complete molecular overview of PARP inhibition. We anticipate that this strategy can be used to integrate functional proteomics with other omics to study molecular processes., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
30. Metabolic Communication by SGLT2 Inhibition.
- Author
-
Billing AM, Kim YC, Gullaksen S, Schrage B, Raabe J, Hutzfeldt A, Demir F, Kovalenko E, Lassé M, Dugourd A, Fallegger R, Klampe B, Jaegers J, Li Q, Kravtsova O, Crespo-Masip M, Palermo A, Fenton RA, Hoxha E, Blankenberg S, Kirchhof P, Huber TB, Laugesen E, Zeller T, Chrysopoulou M, Saez-Rodriguez J, Magnussen C, Eschenhagen T, Staruschenko A, Siuzdak G, Poulsen PL, Schwab C, Cuello F, Vallon V, and Rinschen MM
- Subjects
- Humans, Mice, Animals, Sodium-Glucose Transporter 2 metabolism, Uric Acid, Tryptophan, Proteomics, Uremic Toxins, Glucose, Sodium metabolism, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Diabetes Mellitus, Experimental drug therapy, Diabetes Mellitus, Experimental complications, Induced Pluripotent Stem Cells metabolism, Diabetes Mellitus, Type 2 complications, Cresols, Sulfuric Acid Esters
- Abstract
Background: SGLT2 (sodium-glucose cotransporter 2) inhibitors (SGLT2i) can protect the kidneys and heart, but the underlying mechanism remains poorly understood., Methods: To gain insights on primary effects of SGLT2i that are not confounded by pathophysiologic processes or are secondary to improvement by SGLT2i, we performed an in-depth proteomics, phosphoproteomics, and metabolomics analysis by integrating signatures from multiple metabolic organs and body fluids after 1 week of SGLT2i treatment of nondiabetic as well as diabetic mice with early and uncomplicated hyperglycemia., Results: Kidneys of nondiabetic mice reacted most strongly to SGLT2i in terms of proteomic reconfiguration, including evidence for less early proximal tubule glucotoxicity and a broad downregulation of the apical uptake transport machinery (including sodium, glucose, urate, purine bases, and amino acids), supported by mouse and human SGLT2 interactome studies. SGLT2i affected heart and liver signaling, but more reactive organs included the white adipose tissue, showing more lipolysis, and, particularly, the gut microbiome, with a lower relative abundance of bacteria taxa capable of fermenting phenylalanine and tryptophan to cardiovascular uremic toxins, resulting in lower plasma levels of these compounds (including p-cresol sulfate). SGLT2i was detectable in murine stool samples and its addition to human stool microbiota fermentation recapitulated some murine microbiome findings, suggesting direct inhibition of fermentation of aromatic amino acids and tryptophan. In mice lacking SGLT2 and in patients with decompensated heart failure or diabetes, the SGLT2i likewise reduced circulating p-cresol sulfate, and p-cresol impaired contractility and rhythm in human induced pluripotent stem cell-derived engineered heart tissue., Conclusions: SGLT2i reduced microbiome formation of uremic toxins such as p-cresol sulfate and thereby their body exposure and need for renal detoxification, which, combined with direct kidney effects of SGLT2i, including less proximal tubule glucotoxicity and a broad downregulation of apical transporters (including sodium, amino acid, and urate uptake), provides a metabolic foundation for kidney and cardiovascular protection., Competing Interests: Disclosures Dr Rinschen declares pending research funding from Novo Nordisk unrelated to this work. Over the past 12 months, Dr Vallon has served as a consultant for Lexicon and received speaker honoraria from AstraZeneca and grant support for investigator-initiated research from AstraZeneca, Boehringer Ingelheim, Gilead, Lexicon, Maze, Merck, and Novo-Nordisk. Dr Magnussen receives study-specific funding from the German Center for Cardiovascular Research (DZHK; Promotion of Women Scientists Programme; FKZ 81X3710112), the Deutsche Stiftung für Herzforschung, the Dr Rolf M. Schwiete Stiftung, NDD, and Loewenstein Medical unrelated to the current work. Dr Magnussen has received speaker fees from AstraZeneca, Novartis, Boehringer Ingelheim/Lilly, Bayer, Pfizer, Sanofi, Aventis, Apontis, and Abbott outside this work. Dr Dugourd and R. Fallegger report funding from Pfizer. Dr Saez-Rodriguez reports funding from GSK, Pfizer, and Sanofi and fees from Travere Therapeutics, Stadapharm, and Astex. Dr Hoxha served on advisory boards for Novartis, Morphosys AG, Sotio, and Argenx. The other authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
31. Integrating single-cell multi-omics and prior biological knowledge for a functional characterization of the immune system.
- Author
-
Schäfer PSL, Dimitrov D, Villablanca EJ, and Saez-Rodriguez J
- Subjects
- Cell Communication, Multiomics, Immune System
- Abstract
The immune system comprises diverse specialized cell types that cooperate to defend the host against a wide range of pathogenic threats. Recent advancements in single-cell and spatial multi-omics technologies provide rich information about the molecular state of immune cells. Here, we review how the integration of single-cell and spatial multi-omics data with prior knowledge-gathered from decades of detailed biochemical studies-allows us to obtain functional insights, focusing on gene regulatory processes and cell-cell interactions. We present diverse applications in immunology and critically assess underlying assumptions and limitations. Finally, we offer a perspective on the ongoing technological and algorithmic developments that promise to get us closer to a systemic mechanistic understanding of the immune system., (© 2024. Springer Nature America, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
32. Predicting disease severity in multiple sclerosis using multimodal data and machine learning.
- Author
-
Andorra M, Freire A, Zubizarreta I, de Rosbo NK, Bos SD, Rinas M, Høgestøl EA, de Rodez Benavent SA, Berge T, Brune-Ingebretse S, Ivaldi F, Cellerino M, Pardini M, Vila G, Pulido-Valdeolivas I, Martinez-Lapiscina EH, Llufriu S, Saiz A, Blanco Y, Martinez-Heras E, Solana E, Bäcker-Koduah P, Behrens J, Kuchling J, Asseyer S, Scheel M, Chien C, Zimmermann H, Motamedi S, Kauer-Bonin J, Brandt A, Saez-Rodriguez J, Alexopoulos LG, Paul F, Harbo HF, Shams H, Oksenberg J, Uccelli A, Baeza-Yates R, and Villoslada P
- Subjects
- Humans, Prospective Studies, Leukocytes, Mononuclear, Magnetic Resonance Imaging methods, Patient Acuity, Machine Learning, Multiple Sclerosis diagnostic imaging, Multiple Sclerosis therapy
- Abstract
Background: Multiple sclerosis patients would benefit from machine learning algorithms that integrates clinical, imaging and multimodal biomarkers to define the risk of disease activity., Methods: We have analysed a prospective multi-centric cohort of 322 MS patients and 98 healthy controls from four MS centres, collecting disability scales at baseline and 2 years later. Imaging data included brain MRI and optical coherence tomography, and omics included genotyping, cytomics and phosphoproteomic data from peripheral blood mononuclear cells. Predictors of clinical outcomes were searched using Random Forest algorithms. Assessment of the algorithm performance was conducted in an independent prospective cohort of 271 MS patients from a single centre., Results: We found algorithms for predicting confirmed disability accumulation for the different scales, no evidence of disease activity (NEDA), onset of immunotherapy and the escalation from low- to high-efficacy therapy with intermediate to high-accuracy. This accuracy was achieved for most of the predictors using clinical data alone or in combination with imaging data. Still, in some cases, the addition of omics data slightly increased algorithm performance. Accuracies were comparable in both cohorts., Conclusion: Combining clinical, imaging and omics data with machine learning helps identify MS patients at risk of disability worsening., (© 2023. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
33. Corrigendum to "Post-translational regulation of metabolism in fumarate hydratase deficient cancer cells" [Metabol. Eng. 45 (2018) 149-157].
- Author
-
Gonçalves E, Sciacovelli M, Costa ASH, Tran MGB, Johnson TI, Machado D, Frezza C, and Saez-Rodriguez J
- Published
- 2024
- Full Text
- View/download PDF
34. A network-based transcriptomic landscape of HepG2 cells uncovering causal gene-cytotoxicity interactions underlying drug-induced liver injury.
- Author
-
Wijaya LS, Gabor A, Pot IE, van de Have L, Saez-Rodriguez J, Stevens JL, Le Dévédec SE, Callegaro G, and van de Water B
- Subjects
- Humans, Hep G2 Cells, Gene Expression Profiling, Gene Regulatory Networks, Transcriptome, Chemical and Drug Induced Liver Injury genetics
- Abstract
Drug-induced liver injury (DILI) remains the main reason for drug development attritions largely due to poor mechanistic understanding. Toxicogenomic to interrogate the mechanism of DILI has been broadly performed. Gene coregulation network-based transcriptome analysis is a bioinformatics approach that potentially contributes to improve mechanistic interpretation of toxicogenomic data. Here we performed an extensive concentration time course response-toxicogenomic study in the HepG2 cell line exposed to 20 DILI compounds, 7 reference compounds for stress response pathways, and 10 agonists for cytokines and growth factor receptors. We performed whole transcriptome targeted RNA sequencing to more than 500 conditions and applied weighted gene coregulated network analysis to the transcriptomics data followed by the identification of gene coregulated networks (modules) that were strongly modulated upon the exposure of DILI compounds. Preservation analysis on the module responses of HepG2 and PHH demonstrated highly preserved adaptive stress response gene coregulated networks. We correlated gene coregulated networks with cell death onset and causal relationships of 67 critical target genes of these modules with the onset of cell death was evaluated using RNA interference screening. We identified GTPBP2, HSPA1B, IRF1, SIRT1, and TSC22D3 as essential modulators of DILI compound-induced cell death. These genes were also induced by DILI compounds in PHH. Altogether, we demonstrate the application of large transcriptome datasets combined with network-based analysis and biological validation to uncover the candidate determinants of DILI., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Society of Toxicology.)
- Published
- 2024
- Full Text
- View/download PDF
35. Understanding metric-related pitfalls in image analysis validation.
- Author
-
Reinke A, Tizabi MD, Baumgartner M, Eisenmann M, Heckmann-Nötzel D, Kavur AE, Rädsch T, Sudre CH, Acion L, Antonelli M, Arbel T, Bakas S, Benis A, Blaschko M, Buettner F, Cardoso MJ, Cheplygina V, Chen J, Christodoulou E, Cimini BA, Collins GS, Farahani K, Ferrer L, Galdran A, van Ginneken B, Glocker B, Godau P, Haase R, Hashimoto DA, Hoffman MM, Huisman M, Isensee F, Jannin P, Kahn CE, Kainmueller D, Kainz B, Karargyris A, Karthikesalingam A, Kenngott H, Kleesiek J, Kofler F, Kooi T, Kopp-Schneider A, Kozubek M, Kreshuk A, Kurc T, Landman BA, Litjens G, Madani A, Maier-Hein K, Martel AL, Mattson P, Meijering E, Menze B, Moons KGM, Müller H, Nichyporuk B, Nickel F, Petersen J, Rafelski SM, Rajpoot N, Reyes M, Riegler MA, Rieke N, Saez-Rodriguez J, Sánchez CI, Shetty S, van Smeden M, Summers RM, Taha AA, Tiulpin A, Tsaftaris SA, Calster BV, Varoquaux G, Wiesenfarth M, Yaniv ZR, Jäger PF, and Maier-Hein L
- Abstract
Validation metrics are key for the reliable tracking of scientific progress and for bridging the current chasm between artificial intelligence (AI) research and its translation into practice. However, increasing evidence shows that particularly in image analysis, metrics are often chosen inadequately in relation to the underlying research problem. This could be attributed to a lack of accessibility of metric-related knowledge: While taking into account the individual strengths, weaknesses, and limitations of validation metrics is a critical prerequisite to making educated choices, the relevant knowledge is currently scattered and poorly accessible to individual researchers. Based on a multi-stage Delphi process conducted by a multidisciplinary expert consortium as well as extensive community feedback, the present work provides the first reliable and comprehensive common point of access to information on pitfalls related to validation metrics in image analysis. Focusing on biomedical image analysis but with the potential of transfer to other fields, the addressed pitfalls generalize across application domains and are categorized according to a newly created, domain-agnostic taxonomy. To facilitate comprehension, illustrations and specific examples accompany each pitfall. As a structured body of information accessible to researchers of all levels of expertise, this work enhances global comprehension of a key topic in image analysis validation.
- Published
- 2024
36. Drug-target identification in COVID-19 disease mechanisms using computational systems biology approaches.
- Author
-
Niarakis A, Ostaszewski M, Mazein A, Kuperstein I, Kutmon M, Gillespie ME, Funahashi A, Acencio ML, Hemedan A, Aichem M, Klein K, Czauderna T, Burtscher F, Yamada TG, Hiki Y, Hiroi NF, Hu F, Pham N, Ehrhart F, Willighagen EL, Valdeolivas A, Dugourd A, Messina F, Esteban-Medina M, Peña-Chilet M, Rian K, Soliman S, Aghamiri SS, Puniya BL, Naldi A, Helikar T, Singh V, Fernández MF, Bermudez V, Tsirvouli E, Montagud A, Noël V, Ponce-de-Leon M, Maier D, Bauch A, Gyori BM, Bachman JA, Luna A, Piñero J, Furlong LI, Balaur I, Rougny A, Jarosz Y, Overall RW, Phair R, Perfetto L, Matthews L, Rex DAB, Orlic-Milacic M, Gomez LCM, De Meulder B, Ravel JM, Jassal B, Satagopam V, Wu G, Golebiewski M, Gawron P, Calzone L, Beckmann JS, Evelo CT, D'Eustachio P, Schreiber F, Saez-Rodriguez J, Dopazo J, Kuiper M, Valencia A, Wolkenhauer O, Kitano H, Barillot E, Auffray C, Balling R, and Schneider R
- Subjects
- Humans, SARS-CoV-2, Drug Repositioning, Systems Biology, Computer Simulation, COVID-19
- Abstract
Introduction: The COVID-19 Disease Map project is a large-scale community effort uniting 277 scientists from 130 Institutions around the globe. We use high-quality, mechanistic content describing SARS-CoV-2-host interactions and develop interoperable bioinformatic pipelines for novel target identification and drug repurposing., Methods: Extensive community work allowed an impressive step forward in building interfaces between Systems Biology tools and platforms. Our framework can link biomolecules from omics data analysis and computational modelling to dysregulated pathways in a cell-, tissue- or patient-specific manner. Drug repurposing using text mining and AI-assisted analysis identified potential drugs, chemicals and microRNAs that could target the identified key factors., Results: Results revealed drugs already tested for anti-COVID-19 efficacy, providing a mechanistic context for their mode of action, and drugs already in clinical trials for treating other diseases, never tested against COVID-19., Discussion: The key advance is that the proposed framework is versatile and expandable, offering a significant upgrade in the arsenal for virus-host interactions and other complex pathologies., Competing Interests: AN collaborates with SANOFI-AVENTIS R&D via a public–private partnership grant CIFRE contract, n° 2020/0766. DM and AB are employed at Labvantage-Biomax GmbH and will be affected by any effect of this publication on the commercial version of the AILANI software. JB and BG received consulting fees from Two Six Labs, LLC. TH has served as a shareholder and has consulted for Discovery Collective, Inc. RB and RS are founders and shareholders of MEGENO SA and ITTM SA. JS-R reports funding from GSK, Pfizer and Sanofi and fees/honoraria from Travere Therapeutics, Stadapharm, Astex, Owkin, Pfizer and Grunenthal. JP and LF are employees and shareholders of MedBioinformatics Solutions SL. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be constructed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Niarakis, Ostaszewski, Mazein, Kuperstein, Kutmon, Gillespie, Funahashi, Acencio, Hemedan, Aichem, Klein, Czauderna, Burtscher, Yamada, Hiki, Hiroi, Hu, Pham, Ehrhart, Willighagen, Valdeolivas, Dugourd, Messina, Esteban-Medina, Peña-Chilet, Rian, Soliman, Aghamiri, Puniya, Naldi, Helikar, Singh, Fernández, Bermudez, Tsirvouli, Montagud, Noël, Ponce-de-Leon, Maier, Bauch, Gyori, Bachman, Luna, Piñero, Furlong, Balaur, Rougny, Jarosz, Overall, Phair, Perfetto, Matthews, Rex, Orlic-Milacic, Gomez, De Meulder, Ravel, Jassal, Satagopam, Wu, Golebiewski, Gawron, Calzone, Beckmann, Evelo, D’Eustachio, Schreiber, Saez-Rodriguez, Dopazo, Kuiper, Valencia, Wolkenhauer, Kitano, Barillot, Auffray, Balling, Schneider and the COVID-19 Disease Map Community.)
- Published
- 2024
- Full Text
- View/download PDF
37. Collaborative effect of Csnk1a1 haploinsufficiency and mutant p53 in Myc induction can promote leukemic transformation.
- Author
-
Fuchs SNR, Stalmann USA, Snoeren IAM, Bindels E, Schmitz S, Banjanin B, Hoogenboezem RM, van Herk S, Saad M, Walter W, Haferlach T, Seillier L, Saez-Rodriguez J, Dugourd AJF, Lehmann KV, Ben-Neriah Y, Gleitz HFE, and Schneider RK
- Subjects
- Animals, Humans, Mice, Bone Marrow metabolism, Chromosome Deletion, Haploinsufficiency, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute drug therapy, Myelodysplastic Syndromes genetics
- Abstract
Abstract: It is still not fully understood how genetic haploinsufficiency in del(5q) myelodysplastic syndrome (MDS) contributes to malignant transformation of hematopoietic stem cells. We asked how compound haploinsufficiency for Csnk1a1 and Egr1 in the common deleted region on chromosome 5 affects hematopoietic stem cells. Additionally, Trp53 was disrupted as the most frequently comutated gene in del(5q) MDS using CRISPR/Cas9 editing in hematopoietic progenitors of wild-type (WT), Csnk1a1-/+, Egr1-/+, Csnk1a1/Egr1-/+ mice. A transplantable acute leukemia only developed in the Csnk1a1-/+Trp53-edited recipient. Isolated blasts were indefinitely cultured ex vivo and gave rise to leukemia after transplantation, providing a tool to study disease mechanisms or perform drug screenings. In a small-scale drug screening, the collaborative effect of Csnk1a1 haploinsufficiency and Trp53 sensitized blasts to the CSNK1 inhibitor A51 relative to WT or Csnk1a1 haploinsufficient cells. In vivo, A51 treatment significantly reduced blast counts in Csnk1a1 haploinsufficient/Trp53 acute leukemias and restored hematopoiesis in the bone marrow. Transcriptomics on blasts and their normal counterparts showed that the derived leukemia was driven by MAPK and Myc upregulation downstream of Csnk1a1 haploinsufficiency cooperating with a downregulated p53 axis. A collaborative effect of Csnk1a1 haploinsufficiency and p53 loss on MAPK and Myc upregulation was confirmed on the protein level. Downregulation of Myc protein expression correlated with efficient elimination of blasts in A51 treatment. The "Myc signature" closely resembled the transcriptional profile of patients with del(5q) MDS with TP53 mutation., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
38. Multiscale networks in multiple sclerosis.
- Author
-
Kennedy KE, Kerlero de Rosbo N, Uccelli A, Cellerino M, Ivaldi F, Contini P, De Palma R, Harbo HF, Berge T, Bos SD, Høgestøl EA, Brune-Ingebretsen S, de Rodez Benavent SA, Paul F, Brandt AU, Bäcker-Koduah P, Behrens J, Kuchling J, Asseyer S, Scheel M, Chien C, Zimmermann H, Motamedi S, Kauer-Bonin J, Saez-Rodriguez J, Rinas M, Alexopoulos LG, Andorra M, Llufriu S, Saiz A, Blanco Y, Martinez-Heras E, Solana E, Pulido-Valdeolivas I, Martinez-Lapiscina EH, Garcia-Ojalvo J, and Villoslada P
- Subjects
- Humans, Prospective Studies, Tomography, Optical Coherence methods, Retina, Brain, Heat-Shock Proteins, Multiple Sclerosis
- Abstract
Complex diseases such as Multiple Sclerosis (MS) cover a wide range of biological scales, from genes and proteins to cells and tissues, up to the full organism. In fact, any phenotype for an organism is dictated by the interplay among these scales. We conducted a multilayer network analysis and deep phenotyping with multi-omics data (genomics, phosphoproteomics and cytomics), brain and retinal imaging, and clinical data, obtained from a multicenter prospective cohort of 328 patients and 90 healthy controls. Multilayer networks were constructed using mutual information for topological analysis, and Boolean simulations were constructed using Pearson correlation to identified paths within and among all layers. The path more commonly found from the Boolean simulations connects protein MK03, with total T cells, the thickness of the retinal nerve fiber layer (RNFL), and the walking speed. This path contains nodes involved in protein phosphorylation, glial cell differentiation, and regulation of stress-activated MAPK cascade, among others. Specific paths identified were subsequently analyzed by flow cytometry at the single-cell level. Combinations of several proteins (GSK3AB, HSBP1 or RS6) and immune cells (Th17, Th1 non-classic, CD8, CD8 Treg, CD56 neg, and B memory) were part of the paths explaining the clinical phenotype. The advantage of the path identified from the Boolean simulations is that it connects information about these known biological pathways with the layers at higher scales (retina damage and disability). Overall, the identified paths provide a means to connect the molecular aspects of MS with the overall phenotype., Competing Interests: We have read the journal’s policy and the authors of this manuscript have the following competing interests: KK reports no disclosures. NKdR reports no disclosures. AU received grants and contracts from FISM, Novartis, Biogen, Merck, Fondazione Cariplo, Italian Ministry of Health, received honoraria, or consultation fees from Biogen, Roche, Teva, Merck, Genzyme, Novartis. FI reports no disclosures. MC reports no disclosures. HFH has received honoraria for lecturing or advice from Biogen, Merck, Roche, Novartis and Sanofi. TB has received unrestricted research grants from Biogen and Sanofi-Genzyme. SDB reports no disclosures. EH received honoraria for lecturing and advisory board activity from Biogen, Merck and Sanofi-Genzyme and unrestricted research grant from Merck. SBI reports no disclosures. SAdRB reports no disclosures. FP received honoraria and research support from Alexion, Bayer, Biogen, Chugai, Merck Serono, Novartis, Genzyme, MedImmune, Shire, Teva, and serves on scientific advisory boards for Alexion, MedImmune, and Novartis. He has received funding from Deutsche Forschungsgemeinschaft (DFG Exc 257), Bundesministerium fu?r Bildung und Forschung (Competence Network Multiple Sclerosis), Guthy Jackson Charitable Foundation, EU Framework Program 7, National Multiple Sclerosis Society of the USA. AUB is named as inventor on multiple patents and patents pending owned by Charité - Universitätsmedizin Berlin and/or University of California Irvine for visual computing-based motor function analysis, multiple sclerosis serum biomarkers, and retinal image analysis. He is cofounder and holds shares of Motognosis GmbH and Nocturne GmbH. He serves on the executive board and is Treasurer/Secretary of IMSVISUAL. He received research support from BMWi, BMBF, NIH ICTS, the Kathleen C. Moore Foundation and the Guthy- Jackson Charitable Foundation. Priscilla Ba?cker-Koduah is funded by the DFG Excellence grant to FP (DFG exc 257) and is a Junior scholar of the Einstein Foundation. CC received honoraria for speaking from Bayer and research funding from Novartis, unrelated to this study. SA received a conference grant from Celgene and honoraria for speaking from Alexion, Bayer and Roche. JB reports no disclosures. JSR declares funding from GSK & Sanofi and fees from Travere Therapeutics & Singularity Bio. MR reports no disclosures. LGA is founder and hold stocks at ProtATonce. MA is an employee of Hoffman-La Roche AG, yet this article is related to his activity at the Hospital Clinic of Barcelona. EHML is an employee of the European Medicines Agency (Human Medicines) since 16 April 2019, yet this article is related to her activity at the Hospital Clinic of Barcelona and consequently, it does not in any way represent the views of the Agency or its Committees. SL received compensation for consulting services and speaker honoraria from Biogen Idec, Novartis, TEVA, Genzyme, Sanofi and Merck. AS received compensation for consulting services and speaker honoraria from Bayer-Schering, Merck- Serono, Biogen-Idec, Sanofi-Aventis, TEVA, Novartis and Roche. EMH reports no disclosures. Elisabeth Solana received travel reimbursement from Sanofi and ECTRIMS and reports personal fees from Roche Spain. IPV is currently an employee of UCB pharma, yet this article is related to her activity at the Hospital Clinic of Barcelona. She has received travel reimbursement from Roche Spain and Genzyme-Sanofi, European Academy of Neurology, and European Committee for Treatment and Research in Multiple Sclerosis for international and national meetings over the last 3 years; she holds a patent for an affordable eye-tracking system to measure eye movement in neurologic diseases, and she holds stock in Aura Innovative Robotics. JGO reports no disclosures. PV has received consultancy fees and held stocks from Accure Therapeutics SL, Attune Neurosciences Inc, Spiral Therapeutics Inc, QMenta Inc, CLight Inc, NeuroPrex Inc, StimuSIL and Adhera Health Inc, (Copyright: © 2024 Kennedy et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
39. Metrics reloaded: recommendations for image analysis validation.
- Author
-
Maier-Hein L, Reinke A, Godau P, Tizabi MD, Buettner F, Christodoulou E, Glocker B, Isensee F, Kleesiek J, Kozubek M, Reyes M, Riegler MA, Wiesenfarth M, Kavur AE, Sudre CH, Baumgartner M, Eisenmann M, Heckmann-Nötzel D, Rädsch T, Acion L, Antonelli M, Arbel T, Bakas S, Benis A, Blaschko MB, Cardoso MJ, Cheplygina V, Cimini BA, Collins GS, Farahani K, Ferrer L, Galdran A, van Ginneken B, Haase R, Hashimoto DA, Hoffman MM, Huisman M, Jannin P, Kahn CE, Kainmueller D, Kainz B, Karargyris A, Karthikesalingam A, Kofler F, Kopp-Schneider A, Kreshuk A, Kurc T, Landman BA, Litjens G, Madani A, Maier-Hein K, Martel AL, Mattson P, Meijering E, Menze B, Moons KGM, Müller H, Nichyporuk B, Nickel F, Petersen J, Rajpoot N, Rieke N, Saez-Rodriguez J, Sánchez CI, Shetty S, van Smeden M, Summers RM, Taha AA, Tiulpin A, Tsaftaris SA, Van Calster B, Varoquaux G, and Jäger PF
- Subjects
- Machine Learning, Semantics, Image Processing, Computer-Assisted, Algorithms
- Abstract
Increasing evidence shows that flaws in machine learning (ML) algorithm validation are an underestimated global problem. In biomedical image analysis, chosen performance metrics often do not reflect the domain interest, and thus fail to adequately measure scientific progress and hinder translation of ML techniques into practice. To overcome this, we created Metrics Reloaded, a comprehensive framework guiding researchers in the problem-aware selection of metrics. Developed by a large international consortium in a multistage Delphi process, it is based on the novel concept of a problem fingerprint-a structured representation of the given problem that captures all aspects that are relevant for metric selection, from the domain interest to the properties of the target structure(s), dataset and algorithm output. On the basis of the problem fingerprint, users are guided through the process of choosing and applying appropriate validation metrics while being made aware of potential pitfalls. Metrics Reloaded targets image analysis problems that can be interpreted as classification tasks at image, object or pixel level, namely image-level classification, object detection, semantic segmentation and instance segmentation tasks. To improve the user experience, we implemented the framework in the Metrics Reloaded online tool. Following the convergence of ML methodology across application domains, Metrics Reloaded fosters the convergence of validation methodology. Its applicability is demonstrated for various biomedical use cases., (© 2024. Springer Nature America, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
40. Profiling the heterogeneity of colorectal cancer consensus molecular subtypes using spatial transcriptomics.
- Author
-
Valdeolivas A, Amberg B, Giroud N, Richardson M, Gálvez EJC, Badillo S, Julien-Laferrière A, Túrós D, Voith von Voithenberg L, Wells I, Pesti B, Lo AA, Yángüez E, Das Thakur M, Bscheider M, Sultan M, Kumpesa N, Jacobsen B, Bergauer T, Saez-Rodriguez J, Rottenberg S, Schwalie PC, and Hahn K
- Abstract
The consensus molecular subtypes (CMS) of colorectal cancer (CRC) is the most widely-used gene expression-based classification and has contributed to a better understanding of disease heterogeneity and prognosis. Nevertheless, CMS intratumoral heterogeneity restricts its clinical application, stressing the necessity of further characterizing the composition and architecture of CRC. Here, we used Spatial Transcriptomics (ST) in combination with single-cell RNA sequencing (scRNA-seq) to decipher the spatially resolved cellular and molecular composition of CRC. In addition to mapping the intratumoral heterogeneity of CMS and their microenvironment, we identified cell communication events in the tumor-stroma interface of CMS2 carcinomas. This includes tumor growth-inhibiting as well as -activating signals, such as the potential regulation of the ETV4 transcriptional activity by DCN or the PLAU-PLAUR ligand-receptor interaction. Our study illustrates the potential of ST to resolve CRC molecular heterogeneity and thereby help advance personalized therapy., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
41. Compartments in medulloblastoma with extensive nodularity are connected through differentiation along the granular precursor lineage.
- Author
-
Ghasemi DR, Okonechnikov K, Rademacher A, Tirier S, Maass KK, Schumacher H, Joshi P, Gold MP, Sundheimer J, Statz B, Rifaioglu AS, Bauer K, Schumacher S, Bortolomeazzi M, Giangaspero F, Ernst KJ, Clifford SC, Saez-Rodriguez J, Jones DTW, Kawauchi D, Fraenkel E, Mallm JP, Rippe K, Korshunov A, Pfister SM, and Pajtler KW
- Subjects
- Humans, Cell Differentiation, Disease Progression, Histological Techniques, Medulloblastoma genetics, Cerebellar Neoplasms genetics
- Abstract
Medulloblastomas with extensive nodularity are cerebellar tumors characterized by two distinct compartments and variable disease progression. The mechanisms governing the balance between proliferation and differentiation in MBEN remain poorly understood. Here, we employ a multi-modal single cell transcriptome analysis to dissect this process. In the internodular compartment, we identify proliferating cerebellar granular neuronal precursor-like malignant cells, along with stromal, vascular, and immune cells. In contrast, the nodular compartment comprises postmitotic, neuronally differentiated malignant cells. Both compartments are connected through an intermediate cell stage resembling actively migrating CGNPs. Notably, we also discover astrocytic-like malignant cells, found in proximity to migrating and differentiated cells at the transition zone between the two compartments. Our study sheds light on the spatial tissue organization and its link to the developmental trajectory, resulting in a more benign tumor phenotype. This integrative approach holds promise to explore intercompartmental interactions in other cancers with varying histology., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
42. Spatial transcriptomics of B cell and T cell receptors reveals lymphocyte clonal dynamics.
- Author
-
Engblom C, Thrane K, Lin Q, Andersson A, Toosi H, Chen X, Steiner E, Lu C, Mantovani G, Hagemann-Jensen M, Saarenpää S, Jangard M, Saez-Rodriguez J, Michaëlsson J, Hartman J, Lagergren J, Mold JE, Lundeberg J, and Frisén J
- Subjects
- Humans, Clone Cells metabolism, Gene Expression Profiling methods, B-Lymphocytes metabolism, Pre-B Cell Receptors genetics, Receptors, Antigen, T-Cell genetics, T-Lymphocytes metabolism
- Abstract
The spatial distribution of lymphocyte clones within tissues is critical to their development, selection, and expansion. We have developed spatial transcriptomics of variable, diversity, and joining (VDJ) sequences (Spatial VDJ), a method that maps B cell and T cell receptor sequences in human tissue sections. Spatial VDJ captures lymphocyte clones that match canonical B and T cell distributions and amplifies clonal sequences confirmed by orthogonal methods. We found spatial congruency between paired receptor chains, developed a computational framework to predict receptor pairs, and linked the expansion of distinct B cell clones to different tumor-associated gene expression programs. Spatial VDJ delineates B cell clonal diversity and lineage trajectories within their anatomical niche. Thus, Spatial VDJ captures lymphocyte spatial clonal architecture across tissues, providing a platform to harness clonal sequences for therapy.
- Published
- 2023
- Full Text
- View/download PDF
43. Mapping cardiac remodeling in chronic kidney disease.
- Author
-
Kaesler N, Cheng M, Nagai J, O'Sullivan J, Peisker F, Bindels EMJ, Babler A, Moellmann J, Droste P, Franciosa G, Dugourd A, Saez-Rodriguez J, Neuss S, Lehrke M, Boor P, Goettsch C, Olsen JV, Speer T, Lu TS, Lim K, Floege J, Denby L, Costa I, and Kramann R
- Subjects
- Mice, Animals, Humans, Tumor Necrosis Factor-alpha genetics, Uremic Toxins, Ventricular Remodeling, Renal Insufficiency, Chronic, Heart Failure etiology
- Abstract
Patients with advanced chronic kidney disease (CKD) mostly die from sudden cardiac death and recurrent heart failure. The mechanisms of cardiac remodeling are largely unclear. To dissect molecular and cellular mechanisms of cardiac remodeling in CKD in an unbiased fashion, we performed left ventricular single-nuclear RNA sequencing in two mouse models of CKD. Our data showed a hypertrophic response trajectory of cardiomyocytes with stress signaling and metabolic changes driven by soluble uremia-related factors. We mapped fibroblast to myofibroblast differentiation in this process and identified notable changes in the cardiac vasculature, suggesting inflammation and dysfunction. An integrated analysis of cardiac cellular responses to uremic toxins pointed toward endothelin-1 and methylglyoxal being involved in capillary dysfunction and TNFα driving cardiomyocyte hypertrophy in CKD, which was validated in vitro and in vivo. TNFα inhibition in vivo ameliorated the cardiac phenotype in CKD. Thus, interventional approaches directed against uremic toxins, such as TNFα, hold promise to ameliorate cardiac remodeling in CKD.
- Published
- 2023
- Full Text
- View/download PDF
44. Multicellular factor analysis of single-cell data for a tissue-centric understanding of disease.
- Author
-
Ramirez Flores RO, Lanzer JD, Dimitrov D, Velten B, and Saez-Rodriguez J
- Subjects
- Humans, Gene Expression Profiling, Single-Cell Analysis
- Abstract
Biomedical single-cell atlases describe disease at the cellular level. However, analysis of this data commonly focuses on cell-type-centric pairwise cross-condition comparisons, disregarding the multicellular nature of disease processes. Here, we propose multicellular factor analysis for the unsupervised analysis of samples from cross-condition single-cell atlases and the identification of multicellular programs associated with disease. Our strategy, which repurposes group factor analysis as implemented in multi-omics factor analysis, incorporates the variation of patient samples across cell-types or other tissue-centric features, such as cell compositions or spatial relationships, and enables the joint analysis of multiple patient cohorts, facilitating the integration of atlases. We applied our framework to a collection of acute and chronic human heart failure atlases and described multicellular processes of cardiac remodeling, independent to cellular compositions and their local organization, that were conserved in independent spatial and bulk transcriptomics datasets. In sum, our framework serves as an exploratory tool for unsupervised analysis of cross-condition single-cell atlases and allows for the integration of the measurements of patient cohorts across distinct data modalities., Competing Interests: RR, JL, DD, BV No competing interests declared, JS reports funding from GSK, Pfizer and Sanofi and fees/honoraria from Travere Therapeutics, Stadapharm, Astex, Pfizer and Grunenthal, (© 2023, Ramirez Flores et al.)
- Published
- 2023
- Full Text
- View/download PDF
45. Expanding the coverage of regulons from high-confidence prior knowledge for accurate estimation of transcription factor activities.
- Author
-
Müller-Dott S, Tsirvouli E, Vazquez M, Ramirez Flores RO, Badia-I-Mompel P, Fallegger R, Türei D, Lægreid A, and Saez-Rodriguez J
- Subjects
- Humans, Gene Expression Profiling, Gene Regulatory Networks, Reproducibility of Results, Gene Expression Regulation, Regulon, Transcription Factors metabolism
- Abstract
Gene regulation plays a critical role in the cellular processes that underlie human health and disease. The regulatory relationship between transcription factors (TFs), key regulators of gene expression, and their target genes, the so called TF regulons, can be coupled with computational algorithms to estimate the activity of TFs. However, to interpret these findings accurately, regulons of high reliability and coverage are needed. In this study, we present and evaluate a collection of regulons created using the CollecTRI meta-resource containing signed TF-gene interactions for 1186 TFs. In this context, we introduce a workflow to integrate information from multiple resources and assign the sign of regulation to TF-gene interactions that could be applied to other comprehensive knowledge bases. We find that the signed CollecTRI-derived regulons outperform other public collections of regulatory interactions in accurately inferring changes in TF activities in perturbation experiments. Furthermore, we showcase the value of the regulons by examining TF activity profiles in three different cancer types and exploring TF activities at the level of single-cells. Overall, the CollecTRI-derived TF regulons enable the accurate and comprehensive estimation of TF activities and thereby help to interpret transcriptomics data., (© The Author(s) 2023. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2023
- Full Text
- View/download PDF
46. Enablers and challenges of spatial omics, a melting pot of technologies.
- Author
-
Alexandrov T, Saez-Rodriguez J, and Saka SK
- Subjects
- Metabolomics methods, Computational Biology, Mass Spectrometry, Genomics methods, Proteomics methods
- Abstract
Spatial omics has emerged as a rapidly growing and fruitful field with hundreds of publications presenting novel methods for obtaining spatially resolved information for any omics data type on spatial scales ranging from subcellular to organismal. From a technology development perspective, spatial omics is a highly interdisciplinary field that integrates imaging and omics, spatial and molecular analyses, sequencing and mass spectrometry, and image analysis and bioinformatics. The emergence of this field has not only opened a window into spatial biology, but also created multiple novel opportunities, questions, and challenges for method developers. Here, we provide the perspective of technology developers on what makes the spatial omics field unique. After providing a brief overview of the state of the art, we discuss technological enablers and challenges and present our vision about the future applications and impact of this melting pot., (© 2023 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
47. Gene regulatory network inference in the era of single-cell multi-omics.
- Author
-
Badia-I-Mompel P, Wessels L, Müller-Dott S, Trimbour R, Ramirez Flores RO, Argelaguet R, and Saez-Rodriguez J
- Abstract
The interplay between chromatin, transcription factors and genes generates complex regulatory circuits that can be represented as gene regulatory networks (GRNs). The study of GRNs is useful to understand how cellular identity is established, maintained and disrupted in disease. GRNs can be inferred from experimental data - historically, bulk omics data - and/or from the literature. The advent of single-cell multi-omics technologies has led to the development of novel computational methods that leverage genomic, transcriptomic and chromatin accessibility information to infer GRNs at an unprecedented resolution. Here, we review the key principles of inferring GRNs that encompass transcription factor-gene interactions from transcriptomics and chromatin accessibility data. We focus on the comparison and classification of methods that use single-cell multimodal data. We highlight challenges in GRN inference, in particular with respect to benchmarking, and potential further developments using additional data modalities., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
48. Microbiome-based risk prediction in incident heart failure: a community challenge.
- Author
-
Erawijantari PP, Kartal E, Liñares-Blanco J, Laajala TD, Feldman LE, Carmona-Saez P, Shigdel R, Claesson MJ, Bertelsen RJ, Gomez-Cabrero D, Minot S, Albrecht J, Chung V, Inouye M, Jousilahti P, Schultz JH, Friederich HC, Knight R, Salomaa V, Niiranen T, Havulinna AS, Saez-Rodriguez J, Levinson RT, and Lahti L
- Abstract
Heart failure (HF) is a major public health problem. Early identification of at-risk individuals could allow for interventions that reduce morbidity or mortality. The community-based FINRISK Microbiome DREAM challenge (synapse.org/finrisk) evaluated the use of machine learning approaches on shotgun metagenomics data obtained from fecal samples to predict incident HF risk over 15 years in a population cohort of 7231 Finnish adults (FINRISK 2002, n=559 incident HF cases). Challenge participants used synthetic data for model training and testing. Final models submitted by seven teams were evaluated in the real data. The two highest-scoring models were both based on Cox regression but used different feature selection approaches. We aggregated their predictions to create an ensemble model. Additionally, we refined the models after the DREAM challenge by eliminating phylum information. Models were also evaluated at intermediate timepoints and they predicted 10-year incident HF more accurately than models for 5- or 15-year incidence. We found that bacterial species, especially those linked to inflammation, are predictive of incident HF. This highlights the role of the gut microbiome as a potential driver of inflammation in HF pathophysiology. Our results provide insights into potential modeling strategies of microbiome data in prospective cohort studies. Overall, this study provides evidence that incorporating microbiome information into incident risk models can provide important biological insights into the pathogenesis of HF., Competing Interests: Conflict of Interest Illumina, Inc., and Janssen Pharmaceutica provided additional support by sponsoring the Center for Microbiome Innovation at the University of California San Diego. T.N. has received honoraria for speaking engagements from Servier and AstraZeneca. V.S. has had research collaboration with Bayer AG, unrelated to this study. J.S.-R. has received funding from GSK, Pfizer and Sanofi, and fees/honoraria from Travere Therapeutics, Stadapharm, Astex, Pfizer and Grunenthal. M.I. is a trustee of the Public Health Genomics (PHG) Foundation, a member of the Scientific Advisory Board of Open Targets, and has a research collaboration with AstraZeneca unrelated to this study. R.K. is a cofounder of Micronoma and Biota, holding stock for Gencirq, Cybele, Biomesense, Micronoma, and Biota, serve as a member of the Scientific Advisory Board in Gencirq, DayTwo, Biomesense, and Micronoma and serve as consultant for DayTwo, Cybele, and Biomesense.
- Published
- 2023
- Full Text
- View/download PDF
49. Proteomic Dynamics of Breast Cancer Cell Lines Identifies Potential Therapeutic Protein Targets.
- Author
-
Sun R, Ge W, Zhu Y, Sayad A, Luna A, Lyu M, Liang S, Tobalina L, Rajapakse VN, Yu C, Zhang H, Fang J, Wu F, Xie H, Saez-Rodriguez J, Ying H, Reinhold WC, Sander C, Pommier Y, Neel BG, Aebersold R, and Guo T
- Subjects
- Humans, Proto-Oncogene Proteins c-akt metabolism, Proteomics, Cell Proliferation, Cell Line, Tumor, Drug Resistance, Neoplasm genetics, ErbB Receptors metabolism, Signal Transduction, Triple Negative Breast Neoplasms metabolism
- Abstract
Treatment and relevant targets for breast cancer (BC) remain limited, especially for triple-negative BC (TNBC). We identified 6091 proteins of 76 human BC cell lines using data-independent acquisition (DIA). Integrating our proteomic findings with prior multi-omics datasets, we found that including proteomics data improved drug sensitivity predictions and provided insights into the mechanisms of action. We subsequently profiled the proteomic changes in nine cell lines (five TNBC and four non-TNBC) treated with EGFR/AKT/mTOR inhibitors. In TNBC, metabolism pathways were dysregulated after EGFR/mTOR inhibitor treatment, while RNA modification and cell cycle pathways were affected by AKT inhibitor. This systematic multi-omics and in-depth analysis of the proteome of BC cells can help prioritize potential therapeutic targets and provide insights into adaptive resistance in TNBC., Competing Interests: Conflict of interest T. G. and Y. Z. are shareholders of Westlake Omics Inc. R. A. holds shares in Biognosys. W. G. are employees of Westlake Omics Inc. The remaining authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
50. Democratizing knowledge representation with BioCypher.
- Author
-
Lobentanzer S, Aloy P, Baumbach J, Bohar B, Carey VJ, Charoentong P, Danhauser K, Doğan T, Dreo J, Dunham I, Farr E, Fernandez-Torras A, Gyori BM, Hartung M, Hoyt CT, Klein C, Korcsmaros T, Maier A, Mann M, Ochoa D, Pareja-Lorente E, Popp F, Preusse M, Probul N, Schwikowski B, Sen B, Strauss MT, Turei D, Ulusoy E, Waltemath D, Wodke JAH, and Saez-Rodriguez J
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.