101
|
Cardamone F, Piva A, Löser E, Eichenberger B, Romero-Mulero MC, Zenk F, Shields EJ, Cabezas-Wallscheid N, Bonasio R, Tiana G, Zhan Y, Iovino N. Chromatin landscape at cis-regulatory elements orchestrates cell fate decisions in early embryogenesis. Nat Commun 2025; 16:3007. [PMID: 40148291 PMCID: PMC11950382 DOI: 10.1038/s41467-025-57719-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
The establishment of germ layers during early development is crucial for body formation. The Drosophila zygote serves as a model for investigating these transitions in relation to the chromatin landscape. However, the cellular heterogeneity of the blastoderm embryo poses a challenge for gaining mechanistic insights. Using 10× Multiome, we simultaneously analyzed the in vivo epigenomic and transcriptomic states of wild-type, E(z)-, and CBP-depleted embryos during zygotic genome activation at single-cell resolution. We found that pre-zygotic H3K27me3 safeguards tissue-specific gene expression by modulating cis-regulatory elements. Furthermore, we demonstrate that CBP is essential for cell fate specification functioning as a transcriptional activator by stabilizing transcriptional factors binding at key developmental genes. Surprisingly, while CBP depletion leads to transcriptional arrest, chromatin accessibility continues to progress independently through the retention of stalled RNA Polymerase II. Our study reveals fundamental principles of chromatin-mediated gene regulation essential for establishing and maintaining cellular identities during early embryogenesis.
Collapse
Affiliation(s)
- Francesco Cardamone
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- International Max Planck Research School of Immunobiology, Epigenetics and Metabolism (IMPRS-IEM), Freiburg, Germany
| | - Annamaria Piva
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Eva Löser
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Bastian Eichenberger
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Mari Carmen Romero-Mulero
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Fides Zenk
- Epigenomics of Neurodevelopment, Brain Mind Institute, School of Life Sciences, EPFL - Ecole Polytechnique Federal Lusanne, Ecublens, Switzerland
| | - Emily J Shields
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Urology and Institute of Neuropathology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Nina Cabezas-Wallscheid
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Laboratory of Stem Cell Biology and Ageing, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
- Centre for Integrative Biological Signalling Studies (CIBSS), Freiburg, Germany
| | - Roberto Bonasio
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Guido Tiana
- Università degli Studi di Milano and INFN, Milan, Italy
| | - Yinxiu Zhan
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy.
| | - Nicola Iovino
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
| |
Collapse
|
102
|
Senatore E, Avolio R, Rinaldi L, Chiuso F, Oliva MA, D'Ambrosio C, Bianco AG, Dalla E, Pagnotta SM, Flammia R, Ambrosino C, Memoli D, Turacchio G, Mimoune SI, Toiron Y, Audebert S, Camoin L, Lignitto L, Scaloni A, Arcella A, Feliciello A. Praja2 controls P-body assembly and translation in glioblastoma by non-proteolytic ubiquitylation of DDX6. EMBO Rep 2025:10.1038/s44319-025-00425-5. [PMID: 40148504 DOI: 10.1038/s44319-025-00425-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Glioblastoma multiforme (GBM) is the most lethal form of malignant brain tumor in adults. Dysregulation of protein synthesis contributes to cancer cell plasticity, driving GBM cell heterogeneity, metastatic behavior, and drug resistance. Understanding the complex network and signaling pathways governing protein translation, is therefore an important goal for GBM treatment. Here we identify a novel signaling network centered on the E3 ubiquitin ligase praja2 that controls protein translation in GBM. Praja2 forms a multimeric complex with the RNA helicase DDX6, which inhibits translation of target RNAs within processing bodies (P-bodies). Stimulation of cAMP signaling through activation of G-protein-coupled receptors induces P-body assembly through praja2-mediated non-proteolytic polyubiquitylation of DDX6. Genetic inactivation of praja2 reshapes DDX6/mRNA complexes and translating polysomes and promotes cellular senescence and GBM growth arrest. Expression of an ubiquitylation-defective DDX6 mutant suppresses the assembly of P-bodies and sustains GBM growth. Taken together, our findings identify a cAMP-driven network that controls translation in P-bodies and GBM growth.
Collapse
Affiliation(s)
- Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Rosario Avolio
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | | | - Chiara D'Ambrosio
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici, Naples, Italy
| | - Antonio Giuseppe Bianco
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | - Emiliano Dalla
- Department of Medicine, University of Udine, Udine, Italy
| | | | - Raffaella Flammia
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Concetta Ambrosino
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Domenico Memoli
- Department of Medicine, Surgery and Dentistry SMS, University of Salerno, Salerno, Italy
| | - Gabriele Turacchio
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Sonia Ines Mimoune
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Yves Toiron
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Stephane Audebert
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Luc Camoin
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Luca Lignitto
- Cancer Research Center of Marseille (CRCM), CNRS, Aix Marseille University, INSERM, Institut Paoli-Calmettes, Marseille, France
| | - Andrea Scaloni
- Proteomics, Metabolomics and Mass Spectrometry Laboratory, ISPAAM, National Research Council, Portici, Naples, Italy
| | | | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy.
| |
Collapse
|
103
|
Martinez-Rivas G, Ayala MV, Bender S, Codo GR, Swiderska WK, Lampis A, Pedroza L, Merdanovic M, Sicard P, Pinault E, Richard L, Lavatelli F, Giorgetti S, Canetti D, Rinsant A, Kaaki S, Ory C, Oblet C, Pollet J, Naser E, Carpinteiro A, Roussel M, Javaugue V, Jaccard A, Bonaud A, Delpy L, Ehrmann M, Bridoux F, Sirac C. A mouse model of cardiac immunoglobulin light chain amyloidosis reveals insights into tissue accumulation and toxicity of amyloid fibrils. Nat Commun 2025; 16:2992. [PMID: 40148271 PMCID: PMC11950232 DOI: 10.1038/s41467-025-58307-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Immunoglobulin light chain (LC) amyloidosis (AL) is one of the most common types of systemic amyloidosis but there is no reliable in vivo model for better understanding this disease. Here, we develop a transgenic mouse model producing a human AL LC. We show that the soluble full length LC is not toxic but a single injection of pre-formed amyloid fibrils or an unstable fragment of the LC leads to systemic amyloid deposits associated with early cardiac dysfunction. AL fibrils in mice are highly similar to that of human, arguing for a conserved mechanism of amyloid fibrils formation. Overall, this transgenic mice closely reproduces human cardiac AL amyloidosis and shows that a partial degradation of the LC is likely to initiate the formation of amyloid fibrils in vivo, which in turn leads to cardiac dysfunction. This is a valuable model for research on AL amyloidosis and preclinical evaluation of new therapies.
Collapse
Affiliation(s)
- Gemma Martinez-Rivas
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
| | - Maria Victoria Ayala
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
| | - Sebastien Bender
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
| | - Gilles Roussine Codo
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
| | - Weronika Karolina Swiderska
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
| | - Alessio Lampis
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
| | - Laura Pedroza
- University Duisburg-Essen, Centre for Medical Biotechnology, Essen, Germany
| | - Melisa Merdanovic
- University Duisburg-Essen, Centre for Medical Biotechnology, Essen, Germany
| | - Pierre Sicard
- PhyMedExp, IPAM/Biocampus (IBiSa/France Life Imaging), UMR INSERM 1046-CNRS 9214, universityof Montpellier, Montpellier, France
| | - Emilie Pinault
- BISCEm (Biologie Intégrative Santé Chimie Environnement) Platform, US 42 INSERM/UAR 2015 CNRS, University of Limoges, Limoges, France
| | | | - Francesca Lavatelli
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy
- Research Area, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Sofia Giorgetti
- Department of Molecular Medicine, Institute of Biochemistry, University of Pavia, Pavia, Italy
- Research Area, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Diana Canetti
- Centre for Amyloidosis, Division of Medicine, University College London, London, UK
| | - Alexa Rinsant
- Department of Pathology, University Hospital, Poitiers, France
| | - Sihem Kaaki
- Department of Pathology, University Hospital, Poitiers, France
| | - Cécile Ory
- Department of Pathology, University Hospital, Poitiers, France
| | - Christelle Oblet
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
| | - Justine Pollet
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
| | - Eyad Naser
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Alexander Carpinteiro
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Muriel Roussel
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
| | - Vincent Javaugue
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
- Department of Nephrology, University Hospital, Poitiers, France
| | - Arnaud Jaccard
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
| | - Amélie Bonaud
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
| | - Laurent Delpy
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
| | - Michael Ehrmann
- University Duisburg-Essen, Centre for Medical Biotechnology, Essen, Germany
| | - Frank Bridoux
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France
- Department of Nephrology, University Hospital, Poitiers, France
| | - Christophe Sirac
- CNRS UMR7276/INSERM U1262, University of Limoges, CRIBL lab, team 3 BioPIC, Limoges, France.
- French National Reference Centre for AL Amyloidosis and Other Monoclonal IG Deposition Diseases, University Hospital, Limoges, France.
| |
Collapse
|
104
|
Uzuner Odongo D, İlgün A, Bozkurt FB, Çakır T. A personalized metabolic modelling approach through integrated analysis of RNA-Seq-based genomic variants and gene expression levels in Alzheimer's disease. Commun Biol 2025; 8:502. [PMID: 40148444 PMCID: PMC11950204 DOI: 10.1038/s42003-025-07941-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Generating condition-specific metabolic models by mapping gene expression data to genome-scale metabolic models (GEMs) is a routine approach to elucidate disease mechanisms from a metabolic perspective. On the other hand, integrating variants that perturb enzyme functionality from the same RNA-seq data may enhance GEM accuracy, offering insights into genome-wide metabolic pathology. Our study pioneers the extraction of both transcriptomic and genomic data from the same RNA-seq data to reconstruct personalized metabolic models. We map genes with significantly higher load of pathogenic variants in Alzheimer's disease (AD) onto a human GEM together with the gene expression data. Comparative analysis of the resulting personalized patient metabolic models with the control models shows enhanced accuracy in detecting AD-associated metabolic pathways compared to the case where only expression data is mapped on the GEM. Besides, several otherwise would-be missed pathways are annotated in AD by considering the effect of genomic variants.
Collapse
Affiliation(s)
- Dilara Uzuner Odongo
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Atılay İlgün
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Fatma Betül Bozkurt
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Tunahan Çakır
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey.
| |
Collapse
|
105
|
Qian N, Weinstein JA. Spatial transcriptomic imaging of an intact organism using volumetric DNA microscopy. Nat Biotechnol 2025:10.1038/s41587-025-02613-z. [PMID: 40148595 DOI: 10.1038/s41587-025-02613-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Lymphatic, nervous and tumor tissues exhibit complex physiology arising from three-dimensional interactions within genetically unique microenvironments. Here we develop a technology capable of volumetrically imaging transcriptomes, genotypes and morphologies in a single measurement, without relying on prior knowledge of spatial organization or genetic sequences. Our method extends DNA microscopy into three dimensions at scales involving 107 molecules by forming a distributed intermolecular network of proximal unique DNA barcodes tagging complementary DNA molecules inside the specimen. After sequencing the DNA-encoded network, an image of molecular positions is inferred using geodesic spectral embeddings, a dimensionality reduction approach that we show to be especially suitable for this data-inverse problem. Applying whole-transcriptome volumetric DNA microscopy to intact zebrafish embryos, we demonstrate that three-dimensional image inference recapitulates zebrafish morphology and known gene expression patterns, capturing the spatial organization of gene sequences. Our extension of spatial genetic measurements to three dimensions, independent of prior templates, opens the door to detailed joint resolution of genomics and morphology in biological tissues.
Collapse
Affiliation(s)
- Nianchao Qian
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | - Joshua A Weinstein
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA.
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
106
|
Beddows I, Djirackor S, Omran DK, Jung E, Shih NN, Roy R, Hechmer A, Olshen A, Adelmant G, Tom A, Morrison J, Adams M, Rohrer DC, Schwartz LE, Pearce CL, Auman H, Marto JA, Drescher CW, Drapkin R, Shen H. Impact of BRCA mutations, age, surgical indication, and hormone status on the molecular phenotype of the human Fallopian tube. Nat Commun 2025; 16:2981. [PMID: 40140386 PMCID: PMC11947093 DOI: 10.1038/s41467-025-58145-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
The human Fallopian tube (FT) is an important organ in the female reproductive system and has been implicated as a site of origin for pelvic serous cancers, including high-grade serous tubo-ovarian carcinoma (HGSC). We have generated comprehensive whole-genome bisulfite sequencing, RNA-seq, and proteomic data of over 100 human FTs, with detailed clinical covariate annotations. Our results challenge existing paradigms that extensive epigenetic, transcriptomic and proteomic alterations exist in the FTs from women carrying heterozygous germline BRCA1/2 pathogenic variants. We find minimal differences between BRCA1/2 carriers and non-carriers prior to loss of heterozygosity. Covariates such as age and surgical indication can confound BRCA1/2-related differences reported in the literature, mainly through their impact on cell composition. We systematically document and highlight the degree of variations across normal human FT, defining five groups capturing major cellular and molecular changes across various reproductive stages, pregnancy, and aging. We are able to associate gene, protein, and epigenetic changes with these and other clinical covariates, but not heterozygous BRCA1/2 mutation status. This sheds new light into prevention and early detection of tumorigenesis in populations at high-risk for ovarian cancer.
Collapse
Affiliation(s)
- Ian Beddows
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Svetlana Djirackor
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Dalia K Omran
- Penn Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Euihye Jung
- Penn Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Natalie Nc Shih
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ritu Roy
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Aaron Hechmer
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Adam Olshen
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Guillaume Adelmant
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Blais Proteomics Center, Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ann Tom
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Blais Proteomics Center, Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jacob Morrison
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Marie Adams
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Daniel C Rohrer
- Pathology and Biorepository Core, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Lauren E Schwartz
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Celeste Leigh Pearce
- Department of Epidemiology, University of Michigan School of Public Health and Rogel Cancer Center, Ann Arbor, MI, USA
| | | | - Jarrod A Marto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Blais Proteomics Center, Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Charles W Drescher
- Paul G. Allen Research Center, Swedish Cancer Institute, Seattle, WA, USA.
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| | - Hui Shen
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA.
| |
Collapse
|
107
|
Montandon SA, Beaudier P, Ullate-Agote A, Helleboid PY, Kummrow M, Roig-Puiggros S, Jabaudon D, Andersson L, Milinkovitch MC, Tzika AC. Regulatory and disruptive variants in the CLCN2 gene are associated with modified skin color pattern phenotypes in the corn snake. Genome Biol 2025; 26:73. [PMID: 40140900 PMCID: PMC11948899 DOI: 10.1186/s13059-025-03539-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Snakes exhibit a broad variety of adaptive colors and color patterns, generated by the spatial arrangement of chromatophores, but little is known of the mechanisms responsible for these spectacular traits. Here, we investigate a mono-locus trait with two recessive alleles, motley and stripe, that both cause pattern aberrations in the corn snake. RESULTS We use mapping-by-sequencing to identify the genomic interval where the causal mutations reside. With our differential gene expression analyses, we find that CLCN2 (Chloride Voltage-Gated Channel 2), a gene within the genomic interval, is significantly downregulated in Motley embryonic skin. Furthermore, we identify the stripe allele as the insertion of an LTR-retrotransposon in CLCN2, resulting in a disruptive mutation of the protein. We confirm the involvement of CLCN2 in color pattern formation by producing knock-out snakes that present a phenotype similar to Stripe. In humans and mice, disruption of CLCN2 results in leukoencephalopathy, as well as retinal and testes degeneration. Our single-cell transcriptomic analyses in snakes reveal that CLCN2 is indeed expressed in chromatophores during embryogenesis and in the adult brain, but the behavior and fertility of Motley and Stripe corn snakes are not impacted. CONCLUSIONS Our genomic, transcriptomic, and functional analyses identify a plasma membrane anion channel to be involved in color pattern development in snakes and show that an active LTR-retrotransposon might be a key driver of trait diversification in corn snakes.
Collapse
Affiliation(s)
- Sophie A Montandon
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland
- Present address: Bracco Suisse S.A., Plan-les-Ouates, Switzerland
| | - Pierre Beaudier
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland
| | - Asier Ullate-Agote
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland
- Present address: Biomedical Engineering Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Pierre-Yves Helleboid
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland
| | - Maya Kummrow
- Tierspital, University of Zurich, Zurich, Switzerland
| | - Sergi Roig-Puiggros
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- Clinic of Neurology, Geneva University Hospital, Geneva, Switzerland
| | - Leif Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Michel C Milinkovitch
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland.
| | - Athanasia C Tzika
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
108
|
Deng W, Wang Y, Wang J, Su Y, Li M, Qu K, Wang Y, Li M. Leveraging Vitamin C to Augment Nanoenabled Photothermal Immunotherapy. ACS NANO 2025. [PMID: 40138545 DOI: 10.1021/acsnano.4c17080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Photothermal immunotherapy (PTI) is valuable for precise tumor targeting and immune activation. However, its efficacy is hindered by insufficient immune response, elevated antioxidant levels within tumor, and intrinsic tumor resistance mechanisms. This study introduces Vitamin C (VC), a widely available dietary nutrient, as an effective enhancer for PTI. High-dose VC induces oxidative imbalance in tumor cells, making them more susceptible to nanoenabled near-infrared-II photothermal therapy (NIR-II PTT) with the photosensitizer IR1080. The combination of VC and NIR-II PTT significantly amplifies antitumor immunity by upregulating CXCL16 expression and promoting CXCR6+ T cell infiltration. Clinical data reveal that higher CXCL16 and CXCR6 levels in human tumors correlate with improved survival and T cell infiltration, underscoring the translational potential of this approach. This study positions VC as a safe, accessible, and cost-effective dietary enhancer for PTI, reshaping the role of dietary nutrients in cancer therapy and offering a strategy for overcoming treatment resistance.
Collapse
Affiliation(s)
- Wuxian Deng
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
| | - Yiyuan Wang
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
| | - Junyu Wang
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
| | - Yitan Su
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601 Anhui, China
| | - Mingyang Li
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
| | - Kun Qu
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
| | - Yucai Wang
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601 Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027 Anhui, China
| | - Min Li
- Department of Radiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China
- National Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027 Anhui, China
| |
Collapse
|
109
|
Ducoli L, Zarnegar BJ, Porter DF, Meyers RM, Miao W, Riley NM, Srinivasan S, Jackrazi LV, Yang YY, Li Z, Wang Y, Bertozzi CR, Flynn RA, Khavari PA. irCLIP-RNP and Re-CLIP reveal patterns of dynamic protein assemblies on RNA. Nature 2025:10.1038/s41586-025-08787-5. [PMID: 40140581 DOI: 10.1038/s41586-025-08787-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 02/13/2025] [Indexed: 03/28/2025]
Abstract
RNA-binding proteins (RBPs) control varied processes, including RNA splicing, stability, transport and translation1-3. Dysfunctional RNA-RBP interactions contribute to the pathogenesis of human disease1,4,5; however, characterizing the nature and dynamics of multiprotein assemblies on RNA has been challenging. Here, to address this, non-isotopic ligation-based ultraviolet-light-induced cross-linking and immunoprecipitation6 was combined with mass spectrometry (irCLIP-RNP) to identify RNA-dependent associated proteins (RDAPs) co-bound to RNA with any RBP of interest. irCLIP-RNP defined landscapes of multimeric protein assemblies on RNA, revealing patterns of RBP-RNA associations, including cell-type-selective combinatorial relationships between RDAPs and primary RBPs. irCLIP-RNP also defined dynamic RDAP remodelling in response to epidermal growth factor (EGF), revealing that EGF-induced recruitment of UPF1 adjacent to HNRNPC promotes splicing surveillance of cell proliferation mRNAs. To identify the RNAs simultaneously co-bound by multiple studied RBPs, a sequential immunoprecipitation irCLIP (Re-CLIP) method was also developed. Re-CLIP confirmed binding relationships observed in irCLIP-RNP and identified HNRNPC and UPF1 RBP co-binding on RND3 and DDX3X mRNAs. irCLIP-RNP and Re-CLIP provide a framework to identify and characterize dynamic RNA-protein assemblies in living cells.
Collapse
Affiliation(s)
- Luca Ducoli
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - Brian J Zarnegar
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - Douglas F Porter
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - Robin M Meyers
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - Weili Miao
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - Nicholas M Riley
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Suhas Srinivasan
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | | | - Yen-Yu Yang
- Department of Chemistry, University of California, Riverside, CA, USA
| | - Zhouxian Li
- Department of Chemistry, University of California, Riverside, CA, USA
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, CA, USA
| | - Carolyn R Bertozzi
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Ryan A Flynn
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Paul A Khavari
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA.
- Program in Cancer Biology, Stanford University, Stanford, CA, USA.
- Veterans Affairs, Palo Alto Healthcare System, Palo Alto, CA, USA.
| |
Collapse
|
110
|
Blotenburg M, Suurenbroek L, Bax D, de Visser J, Bhardwaj V, Braccioli L, de Wit E, van Boxtel A, Marks H, Zeller P. Stem cell culture conditions affect in vitro differentiation potential and mouse gastruloid formation. PLoS One 2025; 20:e0317309. [PMID: 40138371 PMCID: PMC11940422 DOI: 10.1371/journal.pone.0317309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 12/24/2024] [Indexed: 03/29/2025] Open
Abstract
Aggregating low numbers of mouse embryonic stem cells (mESCs) and inducing Wnt signalling generates 'gastruloids', self-organising complex structures that display an anteroposterior organisation of cell types derived from all three germ layers. Current gastruloid protocols display considerable heterogeneity between experiments in terms of morphology, elongation efficiency, and cell type composition. We therefore investigated whether altering the mESC pluripotency state would provide more consistent results. By growing three mESC lines from two different genetic backgrounds in different intervals of ESLIF and 2i medium the pluripotency state of cells was modulated, and mESC culture as well as the resulting gastruloids were analysed. Microscopic analysis showed a pre-culture-specific effect on gastruloid formation, in terms of aspect ratio and reproducibility. RNA-seq analysis of the mESC start population confirmed that short-term pulses of 2i and ESLIF modulate the pluripotency state, and result in different cellular states. Since multiple epigenetic regulators were detected among the top differentially expressed genes, we further analysed genome-wide DNA methylation and H3K27me3 distributions. We observed epigenetic differences between conditions, most dominantly in the promoter regions of developmental regulators. Lastly, when we investigated the cell type composition of gastruloids grown from these different pre-cultures, we observed that mESCs subjected to 2i-ESLIF preceding aggregation generated gastruloids more consistently, including more complex mesodermal contributions as compared to the ESLIF-only control. These results indicate that optimisation of the mESCs pluripotency state allows the modulation of cell differentiation during gastruloid formation.
Collapse
Affiliation(s)
- Marloes Blotenburg
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lianne Suurenbroek
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Danique Bax
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Joëlle de Visser
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Vivek Bhardwaj
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Luca Braccioli
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Elzo de Wit
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Antonius van Boxtel
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Hendrik Marks
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Peter Zeller
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
111
|
Scialò C, Zhong W, Jagannath S, Wilkins O, Caredio D, Hruska-Plochan M, Lurati F, Peter M, De Cecco E, Celauro L, Aguzzi A, Legname G, Fratta P, Polymenidou M. Seeded aggregation of TDP-43 induces its loss of function and reveals early pathological signatures. Neuron 2025:S0896-6273(25)00180-1. [PMID: 40157355 DOI: 10.1016/j.neuron.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/24/2024] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Neurodegeneration in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) results from both gain of toxicity and loss of normal function of the RNA-binding protein TDP-43, but their mechanistic connection remains unclear. Increasing evidence suggests that TDP-43 aggregates act as self-templating seeds, propagating pathology through the central nervous system via a prion-like cascade. We developed a robust TDP-43-seeding platform for quantitative assessment of TDP-43 aggregate uptake, cell-to-cell spreading, and loss of function within living cells, while they progress toward pathology. We show that both patient-derived and recombinant TDP-43 pathological aggregates were abundantly internalized by human neuron-like cells, efficiently recruited endogenous TDP-43, and formed cytoplasmic inclusions reminiscent of ALS/FTD pathology. Combining a fluorescent reporter of TDP-43 function with RNA sequencing and proteomics, we demonstrated aberrant cryptic splicing and a loss-of-function profile resulting from TDP-43-templated aggregation. Our data highlight known and novel pathological signatures in the context of seed-induced TDP-43 loss of function.
Collapse
Affiliation(s)
- Carlo Scialò
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Weijia Zhong
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Somanath Jagannath
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Oscar Wilkins
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK; The Francis Crick Institute, London, UK
| | - Davide Caredio
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | | | - Flavio Lurati
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Martina Peter
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Elena De Cecco
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Luigi Celauro
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Giuseppe Legname
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Pietro Fratta
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK; The Francis Crick Institute, London, UK
| | | |
Collapse
|
112
|
Chen X, Han W, Chang X, Tang C, Chen K, Bao L, Zhang L, Hu J, Wang S, Bao Z. High-quality genome assembly of the azooxanthellate coral Tubastraea coccinea (Lesson, 1829). Sci Data 2025; 12:507. [PMID: 40140403 PMCID: PMC11947264 DOI: 10.1038/s41597-025-04839-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Coral reefs are among the most biodiverse and economically significant ecosystems globally, yet they are increasingly degrading due to global climate change and local human activities. The sun coral Tubastraea coccinea (T. coccinea) an obligate heterotroph lacking symbiotic zooxanthellae, exhibits remarkable tolerance to conditions that cause bleaching and mortality in zooxanthellate species. With its extensive low-latitude distribution across multiple oceans, T. coccinea has become a highly invasive species, adversely impacting native species, degrading local ecosystems, and causing significant socio-economic challenges that demand effective management. Despite substantial research efforts, the molecular biology of T. coccinea remains insufficiently characterized. To address this gap, we generated a draft genome assembly for T. coccinea using PacBio Hi-Fi long-read sequencing. The assembly spans 875.9 Mb with a scaffold N50 of 694.3 kb and demonstrates high completeness, with a BUSCO score of 97.4%. A total of 37,307 protein-coding sequences were identified, 95.2% of which were functionally annotated through comparisons with established protein databases. This reference genome provides a valuable resource for understanding the genetic structure of T. coccinea, advancing research into its adaptive mechanism to environmental changes, and informing conservation and management strategies to mitigate its invasive impact.
Collapse
Affiliation(s)
- Xiaomei Chen
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Sanya/Qingdao, China
| | - Wentao Han
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Sanya/Qingdao, China
| | - Xinyao Chang
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Sanya/Qingdao, China
| | - Caiyin Tang
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Sanya/Qingdao, China
| | - Kai Chen
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Lisui Bao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Lingling Zhang
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Sanya/Qingdao, China
| | - Jingjie Hu
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Sanya/Qingdao, China
| | - Shi Wang
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China.
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Sanya/Qingdao, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| | - Zhenmin Bao
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China.
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Sanya/Qingdao, China.
| |
Collapse
|
113
|
Luo L, Yuan F, Palovcak A, Li F, Yuan Q, Calkins D, Manalo Z, Li Y, Wang D, Zhou M, Zhou C, Li M, Tan YD, Bai F, Ban Y, Mason C, Roberts E, Bilbao D, Liu ZJ, Briegel K, Welford SM, Pei XH, Daunert S, Liu W, Zhang Y. Oncogenic properties of wild-type DNA repair gene FANCA in breast cancer. Cell Rep 2025; 44:115480. [PMID: 40146775 DOI: 10.1016/j.celrep.2025.115480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/10/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
FANCA is one of the 23 genes whose deficiencies lead to defective DNA interstrand crosslink repair and cancer-prone Fanconi anemia disease. Beyond its functions in DNA repair and tumor suppression, we report that high FANCA expression is strongly associated with breast cancer development. Overexpression of WT-FANCA significantly promotes breast cancer cell proliferation and tumor growth both in vitro and in vivo, while FANCA deficiency severely compromises the proliferation of breast cancer cells, but not non-tumorigenic breast epithelial cells. Heterozygous knockout of FANCA in breast cancer mouse models is sufficient to cause significant reduction of breast tumor growth in vivo. Furthermore, we have shown that high FANCA expression in breast cancer correlates with promoter hypomethylation in a TET-dependent manner, and TET inhibition recapitulates the proliferation defects caused by FANCA deficiency. Our study identifies the oncogenic properties of WT-FANCA and shows that FANCA is a promising target for breast cancer intervention.
Collapse
Affiliation(s)
- Liang Luo
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Fenghua Yuan
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Anna Palovcak
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Fang Li
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Qingqi Yuan
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daniel Calkins
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Zoe Manalo
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yan Li
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Dazhi Wang
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Mike Zhou
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Catherine Zhou
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Matthew Li
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yuan-De Tan
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Feng Bai
- International Cancer Center, Shenzhen University Medical School, Shenzhen 518060, China
| | - Yuguang Ban
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Christian Mason
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Evan Roberts
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daniel Bilbao
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Pathology & Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Zhao-Jun Liu
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Karoline Briegel
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Scott M Welford
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Xin-Hai Pei
- International Cancer Center, Shenzhen University Medical School, Shenzhen 518060, China
| | - Sylvia Daunert
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Wenjun Liu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Yanbin Zhang
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
114
|
Zhuang QKW, Bauermeister K, Galvez JH, Alogayil N, Batdorj E, de Villena FPM, Taketo T, Bourque G, Naumova AK. Survey of gene, lncRNA and transposon transcription patterns in four mouse organs highlights shared and organ-specific sex-biased regulation. Genome Biol 2025; 26:74. [PMID: 40140847 PMCID: PMC11948892 DOI: 10.1186/s13059-025-03547-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Sex-biased gene regulation is the basis of sexual dimorphism in phenotypes and has been studied across different cell types and different developmental stages. However, sex-biased expression of transposable elements (TEs), which represent nearly half of the mammalian genome and have the potential of influencing genome integrity and regulation, remains underexplored. RESULTS We report a survey of gene, lncRNA, and TE expression in four organs from mice with different combinations of gonadal and genetic sex. The data show remarkable variability among organs with respect to the impact of gonadal sex on transcription with the strongest effects observed in the liver. In contrast, the X-chromosome dosage alone had a modest influence on sex-biased transcription across organs, albeit interaction between X-dosage and gonadal sex cannot be ruled out. The presence of the Y-chromosome influences TE, but not gene or lncRNA, expression in the liver. Notably, 90% of sex-biased TEs (sDETEs) reside in clusters. Moreover, 54% of these clusters overlap or reside less than 100 kb from sex-biased genes or lncRNAs, share the same sex bias, and also have higher expression levels than sDETE clusters that do not co-localize with other types of sex-biased transcripts. We test the heterochromatic sink hypothesis that predicts higher expression of TEs in XX individuals finding no evidence to support it. CONCLUSIONS Our data show that sex-biased expression of TEs varies among organs with the highest numbers of sDETEs found in the liver following trends observed for genes and lncRNAs. It is enhanced by proximity to other types of sex-biased transcripts.
Collapse
Affiliation(s)
- Qinwei Kim-Wee Zhuang
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1C7, Canada
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto, 606-8303, Japan
| | - Klara Bauermeister
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1C7, Canada
| | - Jose Hector Galvez
- Canadian Centre for Computational Genomics, Montreal, QC, H3A 0G1, Canada
| | - Najla Alogayil
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1C7, Canada
| | - Enkhjin Batdorj
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1C7, Canada
| | - Fernando Pardo Manuel de Villena
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Teruko Taketo
- The Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
- Department of Surgery, McGill University, Montreal, QC, H4A 3J1, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, H4A 3J1, Canada
| | - Guillaume Bourque
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1C7, Canada.
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto, 606-8303, Japan.
- Canadian Centre for Computational Genomics, Montreal, QC, H3A 0G1, Canada.
| | - Anna K Naumova
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1C7, Canada.
- The Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada.
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
115
|
Sabikunnahar B, Snyder JP, Rodriguez PD, Sessions KJ, Amiel E, Frietze SE, Krementsov DN. Natural genetic variation in wild-derived mice controls host survival and transcriptional responses during endotoxic shock. Immunohorizons 2025; 9:vlaf007. [PMID: 40139977 PMCID: PMC11945298 DOI: 10.1093/immhor/vlaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/22/2025] [Indexed: 03/29/2025] Open
Abstract
Innate immune cells sense microbial danger signals, resulting in dynamic transcriptional reprogramming and rapid inflammatory responses. If not properly regulated, such responses can be detrimental to the host, as is seen in septic shock. A better understanding of the genetic regulation of responses during endotoxemia could provide potential therapeutic insights. However, the majority of animal model studies have been performed using classic inbred laboratory strains of mice, capturing limited genetic diversity. Here, we compared classic inbred C57BL/6 (B6) mice with wild-derived and genetically divergent PWD/PhJ (PWD) mice using in vivo and in vitro models of endotoxemia. Compared with B6 mice, PWD mice were markedly resistant to bacterial lipopolysaccharide (LPS)-induced endotoxic shock. Using LPS stimulation of bone marrow derived dendritic cells (BMDC) and RNA sequencing, we demonstrate that B6 and PWD BMDCs exhibit partially overlapping yet highly divergent transcriptional responses, with B6 skewed toward stereotypical proinflammatory pathway activation, and PWD engaging regulatory or developmental pathways. To dissect genetic regulation of inflammatory responses by allelic variants, we used BMDCs from a sub-consomic strain carrying a ∼50 Mb PWD-derived portion of chromosome 11 on the B6 background. This identified a subset of cis-regulated and a large number of trans-regulated genes. Bioinformatic analyses identified candidate trans regulators encoded in the chromosome 11 locus as transcription factors Irf1, Ncor1, and Srebf1. Our results demonstrate that natural genetic variation controls host survival and transcriptional reprogramming during endotoxemia, suggesting possibilities for prediction of sepsis risk and/or personalized therapeutic interventions.
Collapse
Affiliation(s)
- Bristy Sabikunnahar
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Julia P Snyder
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Princess D Rodriguez
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT, United States
| | - Katherine J Sessions
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Eyal Amiel
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Seth E Frietze
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Dimitry N Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| |
Collapse
|
116
|
Agarwal S, Taft E, Gauthier M, Darcy J, Buckowing K, Berman D, Davis WP, Rogers AB, Janas MM. Mechanistic Insights into Hybridization-Based Off-Target Activity of GalNAc-siRNA Conjugates. Nucleic Acid Ther 2025. [PMID: 40134378 DOI: 10.1089/nat.2024.0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025] Open
Abstract
Nonclinical safety screening of small interfering RNAs (siRNAs) conjugated to a trivalent N-acetylgalactosamine (GalNAc) ligand is typically carried out in rats at exaggerated exposures in a repeat-dose regimen. We have previously shown that at these suprapharmacological doses, hepatotoxicity observed with a subset of GalNAc-siRNAs is largely driven by undesired RNA-induced silencing complex (RISC)-mediated antisense strand seed-based off-target activity, similar to microRNA-like regulation. However, the RISC component requirements for off-target activity of siRNAs have not been evaluated. Here, we evaluate the roles of major RISC components, AGO and TNRC6 (or GW182) proteins, in driving on- and off-target activity of GalNAc-siRNAs in hepatocytes, in vitro and in vivo. We demonstrate that knocking down AGO2, but not AGO1 or AGO4, is protective against GalNAc-siRNA-driven off-target activity and hepatotoxicity. As expected, knocking down AGO2, but not AGO1 or AGO4, reduces the on-target activity of GalNAc-siRNA. Similarly, knocking down TNRC6 paralogs, TNRC6A or TNRC6B, but not TNRC6C, is protective against off-target activity and hepatotoxicity while having minimal impact on the on-target activity of GalNAc-siRNA. These data indicate that while AGO2 is the only RISC component required for the on-target activity of GalNAc-siRNAs, the undesired off-target activity and hepatotoxicity of a subset of GalNAc-siRNAs are mediated via the RISC composed predominantly of AGO2 and TNRC6 paralogs TNRC6A and/or TNRC6B.
Collapse
Affiliation(s)
- Saket Agarwal
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | | | | | - Justin Darcy
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | | | - Daniel Berman
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| | | | | | - Maja M Janas
- Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA
| |
Collapse
|
117
|
Megat S, Marques C, Hernán-Godoy M, Sellier C, Stuart-Lopez G, Dirrig-Grosch S, Gorin C, Brunet A, Fischer M, Keime C, Kessler P, Mendoza-Parra MA, Zwamborn RAJ, Veldink JH, Scholz SW, Ferrucci L, Ludolph A, Traynor B, Chio A, Dupuis L, Rouaux C. CREB3 gain of function variants protect against ALS. Nat Commun 2025; 16:2942. [PMID: 40140376 PMCID: PMC11947196 DOI: 10.1038/s41467-025-58098-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly evolving neurodegenerative disease arising from the loss of glutamatergic corticospinal neurons (CSN) and cholinergic motoneurons (MN). Here, we performed comparative cross-species transcriptomics of CSN using published snRNA-seq data from the motor cortex of ALS and control postmortem tissues, and performed longitudinal RNA-seq on CSN purified from male Sod1G86R mice. We report that CSN undergo ER stress and altered mRNA translation, and identify the transcription factor CREB3 and its regulatory network as a resilience marker of ALS, not only amongst vulnerable neuronal populations, but across all neuronal populations as well as other cell types. Using genetic and epidemiologic analyses we further identify the rare variant CREB3R119G (rs11538707) as a positive disease modifier in ALS. Through gain of function, CREB3R119G decreases the risk of developing ALS and the motor progression rate of ALS patients.
Collapse
Affiliation(s)
- Salim Megat
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France.
| | - Christine Marques
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Marina Hernán-Godoy
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Chantal Sellier
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Geoffrey Stuart-Lopez
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Sylvie Dirrig-Grosch
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Charlotte Gorin
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Aurore Brunet
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Mathieu Fischer
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Céline Keime
- Université de Strasbourg, Inserm UMR-S 1258, CNRS UMR-S 7104, Institut de Génétique, Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, France
| | - Pascal Kessler
- Université de Strasbourg, Inserm UMS 38, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Marco Antonio Mendoza-Parra
- UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Evry-val-d'Essonne, University Paris-Saclay, Evry, France
| | - Ramona A J Zwamborn
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Jan H Veldink
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Luigi Ferrucci
- Intramural Research Program of the National Institute on Aging, NIH, Baltimore, MD, USA
| | | | - Bryan Traynor
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Adriano Chio
- ALS Center "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy
| | - Luc Dupuis
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Caroline Rouaux
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France.
| |
Collapse
|
118
|
Decoene I, Svitina H, Belal Hamed M, Economou A, Stegen S, Luyten FP, Papantoniou I. Callus organoids reveal distinct cartilage to bone transition mechanisms across donors and a role for biological sex. Bone Res 2025; 13:41. [PMID: 40140357 PMCID: PMC11947321 DOI: 10.1038/s41413-025-00418-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/29/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Clinical translation of tissue-engineered advanced therapeutic medicinal products is hindered by a lack of patient-dependent and independent in-process biological quality controls that are reflective of in vivo outcomes. Recent insights into the mechanism of native bone repair highlight a robust path dependence. Organoid-based bottom-up developmental engineering mimics this path-dependence to design personalized living implants scaffold-free, with in-build outcome predictability. Yet, adequate (noninvasive) quality metrics of engineered tissues are lacking. Moreover, insufficient insight into the role of donor variability and biological sex as influencing factors for the mechanism toward bone repair hinders the implementation of such protocols for personalized bone implants. Here, male and female bone-forming organoids were compared to non-bone-forming organoids regarding their extracellular matrix composition, transcriptome, and secreted proteome signatures to directly link in vivo outcomes to quality metrics. As a result, donor variability in bone-forming callus organoids pointed towards two distinct pathways to bone, through either a hypertrophic cartilage or a fibrocartilaginous template. The followed pathway was determined early, as a biological sex-dependent activation of distinct progenitor populations. Independent of donor or biological sex, a cartilage-to-bone transition was driven by a common panel of secreted factors that played a role in extracellular matrix remodeling, mineralization, and attraction of vasculature. Hence, the secreted proteome is a source of noninvasive biomarkers that report on biological potency and could be the missing link toward data-driven decision-making in organoid-based bone tissue engineering.
Collapse
Affiliation(s)
- Isaak Decoene
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
- Prometheus Translational Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
| | - Hanna Svitina
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
- Prometheus Translational Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
| | - Mohamed Belal Hamed
- Laboratory of Molecular Bacteriology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Molecular Biology Department, National Research Centre, 33 El Buhouth st, Dokii, 12622, Cairo, Egypt
- Department of Neurosciences, Leuven Research Institute for Neuroscience and Disease (LIND), KU Leuven, VIB-KU Leuven Center for Brain & Disease Research, Herestraat, 3000, Leuven, Belgium
| | - Anastassios Economou
- Laboratory of Molecular Bacteriology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Steve Stegen
- Prometheus Translational Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Frank P Luyten
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
- Prometheus Translational Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium
| | - Ioannis Papantoniou
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium.
- Prometheus Translational Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, box 813, 3000, Leuven, Belgium.
- Institute for Chemical Engineering Sciences, Foundation for Research and Technology-Hellas (FORTH), Stadiou Street, Platani, box 1414, 26504, Patras, Greece.
| |
Collapse
|
119
|
Zapata-Linares N, Toillon I, Wanherdrick K, Pigenet A, Duhalde F, Binvignat M, Martin-Uriz PS, Louvet L, Calleja-Cervantes ME, Ghali Mhenni O, Guibert C, Nourissat G, Nogier A, Leterme D, Broux O, Magneron P, Prosper F, Chauveau C, Landoulsi J, Berenbaum F, Rodriguez-Madoz JR, Lafage-Proust MH, Lucas S, Houard X. Implication of bone marrow adipose tissue in bone homeostasis during osteoarthritis. Osteoarthritis Cartilage 2025:S1063-4584(25)00870-2. [PMID: 40154729 DOI: 10.1016/j.joca.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 03/16/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVE To explore the role of bone marrow adipocytes (BMAds) in osteoarthritis (OA). METHODS Male and female C57BL/6 mice (n=4/group) underwent meniscectomy (MNX) or SHAM surgery. OA was determined using OARSI score and the number of perilipin+ adipocytes was quantified. Mesenchymal Stromal Cells (MSCs) from MNX and SHAM mice were differentiated into osteoblasts and adipocytes. Human adipocytes and MSCs (n=8) were enzymatically isolated from epiphyseal and metaphyseal marrow, and from subcutaneous adipose tissue (SCAT) of hip OA patients. Human OA MSCs were differentiated into osteoblasts and adipocytes (OA-Diff-hAdipo). Gene expression patterns of epiphyseal and metaphyseal BMAds, SCAT adipocytes and OA-Diff-hAdipo were evaluated by RNAseq (n=4). The effect conditioned media from OA epiphyseal bone (n=5) on the alkaline phosphatase (ALP) activity and mineralization kinetics was assessed in vitro. RESULTS Increase in BMAd density was positively correlated with cartilage degradation in MNX mice. OA modified the differentiation capacity of MSCs, accelerating adipocyte differentiation and failing to produce osteoblasts in both human and mice. Human epiphyseal, metaphyseal and SCAT adipocytes from the same OA patients each displayed a specific transcriptome, suggesting different functions. Enrichment analysis defined metaphyseal OA-BMAds as cells implicated in hematopoietic stem cell differentiation. On the other hand, epiphyseal OA-BMAds were considered as osteogenic cells showing an up-regulation of genes related to bone mineralization and remodeling. Specifically, OA epiphysis-secreted molecules decreased ALP activity and altered in vitro the mineralization process. CONCLUSION All these results support the emergence of BMAds as new cell partners in OA, opening new venues for therapeutic approaches.
Collapse
Affiliation(s)
- Natalia Zapata-Linares
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Indira Toillon
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Kristell Wanherdrick
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Audrey Pigenet
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Fanny Duhalde
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France; Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005, Paris, France
| | - Marie Binvignat
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | | | - Loïc Louvet
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille F-59000 Lille, CHU Lille F-59000 Lille, France
| | - Maria E Calleja-Cervantes
- Hemato-Oncology Program. CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain; Computational Biology Program. CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain
| | - Olfa Ghali Mhenni
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille F-59000 Lille, CHU Lille F-59000 Lille, France
| | - Clément Guibert
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005, Paris, France
| | - Geoffroy Nourissat
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | | | - Damien Leterme
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille F-59000 Lille, CHU Lille F-59000 Lille, France
| | - Odile Broux
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille F-59000 Lille, CHU Lille F-59000 Lille, France
| | - Paul Magneron
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Felipe Prosper
- Hemato-Oncology Program. CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC). Madrid, Spain; Hematology and Cell Therapy Department. Clinica Universidad de Navarra, IdiSNA. Pamplona, Spain; Cancer Center Universidad de Navarra (CCUN). Pamplona, Spain
| | - Christophe Chauveau
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille F-59000 Lille, CHU Lille F-59000 Lille, France
| | - Jessem Landoulsi
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005, Paris, France
| | - Francis Berenbaum
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France; heumatology Department, AP-HP Saint-Antoine Hospital, 184, rue du Faubourg Saint-Antoine, 75012, Paris, France
| | - Juan R Rodriguez-Madoz
- Hemato-Oncology Program. CIMA Universidad de Navarra-IdiSNA, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC). Madrid, Spain; Cancer Center Universidad de Navarra (CCUN). Pamplona, Spain
| | - Marie-Hélène Lafage-Proust
- Université de Lyon - Université Jean Monnet, INSERM U1059, Faculté de Médecine, F-42270 Saint-Priest en Jarez, France
| | - Stéphanie Lucas
- Marrow Adiposity and Bone Lab (MABLab) ULR4490, Université du Littoral Côte d'Opale, F-62200 Boulogne sur Mer, Univ. Lille F-59000 Lille, CHU Lille F-59000 Lille, France
| | - Xavier Houard
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| |
Collapse
|
120
|
Zhao X, Liu Y, Ye X, Wang F, Fang Q, Ye G. Chromosome-level genome assembly of an endoparasitoid Cotesia ruficrus. Sci Data 2025; 12:508. [PMID: 40140407 PMCID: PMC11947152 DOI: 10.1038/s41597-025-04837-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Cotesia ruficrus is a gregarious larval endoparasitoid of many important agricultural pest moths, such as the fall armyworm, northern armyworm and rice leaffolder. While eight of Cotesia genomes had been reported, high-quality genomic data of C. ruficrus has yet to be performed. This study presents a chromosome-scale genome assembly of C. ruficrus, utilizing both PacBio HiFi-reads and Hi-C reads. The genome size is about 185.3 Mb harboring 10 chromosomes, and 209 unanchored scaffolds with a scaffold N50 length of 15.81 Mb. The anchored rate of sequences to chromosomes is 95.55%. A total of 14,001 genes have been annotated. This work delivers a first high-quality, chromosome-scale genome for C. ruficrus, enriching the datasets for comparative evolutionary studies within the Cotesia genus. Additionally, it offers a valuable genomic resource for future biological and genetic research, as well as for enhancing pest management strategies, such as biological control.
Collapse
Affiliation(s)
- Xianxin Zhao
- State Key Laboratory of Rice Biology and Breeding & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, Zhejiang University, Hangzhou, China
| | - Yuanyuan Liu
- State Key Laboratory of Rice Biology and Breeding & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, Zhejiang University, Hangzhou, China
| | - Xinhai Ye
- College of Advanced Agriculture Sciences, Zhejiang A&F University, Hangzhou, China
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Zhejiang A&F University, Hangzhou, 311300, China
| | - Fang Wang
- State Key Laboratory of Rice Biology and Breeding & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, Zhejiang University, Hangzhou, China
| | - Qi Fang
- State Key Laboratory of Rice Biology and Breeding & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, Zhejiang University, Hangzhou, China
| | - Gongyin Ye
- State Key Laboratory of Rice Biology and Breeding & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
121
|
Nakagawa T, Luebeck J, Zhu K, Lange JT, Sasik R, Phillips C, Sadat S, Javadzadeh S, Yang Q, Monther A, Fassardi S, Wang A, Pestonjamasp K, Rosenthal B, Fisch KM, Mischel P, Bafna V, Califano JA. Inhibition of human-HPV hybrid ecDNA enhancers reduces oncogene expression and tumor growth in oropharyngeal cancer. Nat Commun 2025; 16:2964. [PMID: 40140353 PMCID: PMC11947173 DOI: 10.1038/s41467-025-57447-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 02/22/2025] [Indexed: 03/28/2025] Open
Abstract
Extrachromosomal circular DNA (ecDNA) has been found in most types of human cancers, and ecDNA incorporating viral genomes has recently been described, specifically in human papillomavirus (HPV)-mediated oropharyngeal cancer (OPC). However, the molecular mechanisms of human-viral hybrid ecDNA (hybrid ecDNA) for carcinogenesis remains elusive. We characterize the epigenetic status of hybrid ecDNA using HPVOPC cell lines and patient-derived tumor xenografts, identifying HPV oncogenes E6/E7 in hybrid ecDNA are flanked by previously unrecognized somatic DNA enhancers and HPV L1 enhancers, with strong cis-interactions. Targeting of these enhancers by clustered regularly interspaced short palindromic repeats interference or hybrid ecDNA by bromodomain and extra-terminal inhibitor reduces E6/E7 expression, and significantly inhibites in vitro and/or in vivo growth only in ecDNA(+) models. HPV DNA in hybrid ecDNA structures are associated with previously unrecognized somatic and HPV enhancers in hybrid ecDNA that drive HPV ongogene expression and carcinogenesis, and can be targeted with ecDNA disrupting therapeutics.
Collapse
Affiliation(s)
- Takuya Nakagawa
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
- Department of Otorhinolaryngology, Head and Neck Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan.
- Health and Disease Omics Center, Chiba University, Chiba, Japan.
| | - Jens Luebeck
- Department of Computer Science and Engineering, UC San Diego, La Jolla, CA, USA
| | - Kaiyuan Zhu
- Department of Computer Science and Engineering, UC San Diego, La Jolla, CA, USA
| | - Joshua T Lange
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Roman Sasik
- Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Chad Phillips
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Sayed Sadat
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Sara Javadzadeh
- Department of Computer Science and Engineering, UC San Diego, La Jolla, CA, USA
| | - Qian Yang
- Department of Cellular and Molecular Medicine, Center for Epigenomics, University of California, San Diego, La Jolla, CA, USA
| | - Abdula Monther
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Santiago Fassardi
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Allen Wang
- Department of Cellular and Molecular Medicine, Center for Epigenomics, University of California, San Diego, La Jolla, CA, USA
| | - Kersi Pestonjamasp
- Cancer Center Microscopy Core, University of California, San Diego, La Jolla, CA, USA
| | - Brin Rosenthal
- Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Kathleen M Fisch
- Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Paul Mischel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Vineet Bafna
- Department of Computer Science and Engineering, UC San Diego, La Jolla, CA, USA
| | - Joseph A Califano
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
- Department of Otolaryngology-Head and Neck Surgery and Gleiberman Head and Neck Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
122
|
Lee S, Barbour JA, Tam YM, Yang H, Huang Y, Wong JWH. LocusMasterTE: integrating long-read RNA sequencing improves locus-specific quantification of transposable element expression. Genome Biol 2025; 26:72. [PMID: 40140852 PMCID: PMC11948968 DOI: 10.1186/s13059-025-03522-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Transposable elements (TEs) can influence human diseases by disrupting genome integrity, yet their quantification has been challenging due to the repetitive nature of these sequences across the genome. We develop LocusMasterTE, a method that integrates long-read with short-read RNA-seq to increase the accuracy of TE expression quantification. By incorporating fractional transcript per million values from long-read sequencing data into an expectation-maximization algorithm, LocusMasterTE reassigns multi-mapped reads, enhancing accuracy in short-read-based TE quantification. We validate the method with simulated and human datasets. LocusMasterTE may give new insights into TE functions through precise quantification.
Collapse
Affiliation(s)
- Sojung Lee
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Jayne A Barbour
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Yee Man Tam
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Haocheng Yang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Yuanhua Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
- Center for Translational Stem Cell Biology, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Jason W H Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China.
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China.
| |
Collapse
|
123
|
Gerakopoulos V, Ramos C, Müller C, Walterskirchen N, Vintila S, Zotter C, Ilg M, Pap A, Riss S, Bergmann M, Unger LW, Vogt AB, Oehler R, Lukowski SW. Single-cell transcriptomic analysis identifies tissue-specific fibroblasts as the main modulators of myeloid cells in peritoneal metastasis of different origin. Cancer Lett 2025:217678. [PMID: 40154914 DOI: 10.1016/j.canlet.2025.217678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Colorectal cancer (CRC) peritoneal metastasis (CPM) is related to limited therapy options and poor prognosis. Although stromal cells heavily infiltrate most CPMs, interactions between different cell types in their microenvironment remain unclear. Here, we investigated tumor and distant normal tissue from CPM and CRC patients using single-cell RNA sequencing. Investigating the incoming and outgoing signals between cells revealed that fibroblasts dominate the CPM signaling landscape with myeloid cells as their strongest interaction partner. Using immunohistochemistry, we confirmed that fibroblasts co-localize with macrophages in the CPM microenvironment. A fibroblast sub-population detected only in CPM and normal peritoneum demonstrated immunoregulatory properties in co-culture experiments, and was further detected in additional peritoneal malignancies derived from ovarian and gastric origin. This novel fibroblast type and its communication with macrophages could be attractive targets for therapeutic interventions in CPM and potentially peritoneal surface malignancies in general.
Collapse
Affiliation(s)
- Vasileios Gerakopoulos
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Cristiano Ramos
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Catharina Müller
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Natalie Walterskirchen
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Stefania Vintila
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Chiara Zotter
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Mathias Ilg
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120 Vienna, Austria
| | - Anna Pap
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120 Vienna, Austria
| | - Stefan Riss
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Bergmann
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Lukas W Unger
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria; Dept. of Colorectal Surgery, Oxford University Hospitals, Old Rd, Headington, Oxford OX3 7LE, United Kingdom
| | - Anne B Vogt
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120 Vienna, Austria
| | - Rudolf Oehler
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria.
| | - Samuel W Lukowski
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120 Vienna, Austria
| |
Collapse
|
124
|
John K, Huntress I, Smith E, Chou H, Tollison TS, Covarrubias S, Crisci E, Carpenter S, Peng X. Human long noncoding RNA VILMIR is induced by major respiratory viral infections and modulates the host interferon response. J Virol 2025:e0014125. [PMID: 40130878 DOI: 10.1128/jvi.00141-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) are a newer class of noncoding transcripts identified as key regulators of biological processes. Here, we aimed to identify novel lncRNA targets that play critical roles in major human respiratory viral infections by systematically mining large-scale transcriptomic data sets. Using bulk RNA-sequencing (RNA-seq) analysis, we identified a previously uncharacterized lncRNA, named virus-inducible lncRNA modulator of interferon response (VILMIR), that was consistently upregulated after in vitro influenza infection across multiple human epithelial cell lines and influenza A virus subtypes. VILMIR was also upregulated after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and respiratory syncytial virus (RSV) infections in vitro. We experimentally confirmed the response of VILMIR to influenza infection and interferon-beta (IFN-β) treatment in the A549 human epithelial cell line and found the expression of VILMIR was robustly induced by IFN-β treatment in a dose- and time-specific manner. Single-cell RNA-seq analysis of bronchoalveolar lavage fluid samples from coronavirus disease 2019 (COVID-19) patients uncovered that VILMIR was upregulated across various cell types, including at least five immune cells. The upregulation of VILMIR in immune cells was further confirmed in the human T cell and monocyte cell lines, SUP-T1 and THP-1, after IFN-β treatment. Finally, we found that knockdown of VILMIR expression reduced the magnitude of host transcriptional responses to both IFN-β treatment and influenza A virus infection in A549 cells. Together, our results show that VILMIR is a novel interferon-stimulated gene (ISG) that regulates the host interferon response and may be a potential therapeutic target for human respiratory viral infections upon further mechanistic investigation.IMPORTANCEIdentifying host factors that regulate the immune response to human respiratory viral infection is critical to developing new therapeutics. Human long noncoding RNAs (lncRNAs) have been found to play key regulatory roles during biological processes; however, the majority of lncRNA functions within the host antiviral response remain unknown. In this study, we identified that a previously uncharacterized lncRNA, virus-inducible lncRNA modulator of interferon response (VILMIR), is upregulated after major respiratory viral infections including influenza, severe acute respiratory syndrome coronavirus 2, and respiratory syncytial virus. We demonstrated that VILMIR is an interferon-stimulated gene that is upregulated after interferon-beta (IFN-β) in several human cell types. We also found that knockdown of VILMIR reduced the magnitude of host transcriptional responses to IFN-β treatment and influenza A infection in human epithelial cells. Our results reveal that VILMIR regulates the host interferon response and may present a new therapeutic target during human respiratory viral infections.
Collapse
Affiliation(s)
- Kristen John
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Genetics & Genomics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Ian Huntress
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Ethan Smith
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Hsuan Chou
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Tammy S Tollison
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Sergio Covarrubias
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, USA
| | - Elisa Crisci
- Department of Population Health and Pathobiology, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Susan Carpenter
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, USA
| | - Xinxia Peng
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
125
|
Cai L, Li B, Zhou Q, Du J, Yang W, Shen L, He C. N-acetyltransferase 10 catalyzes RNA N 4-acetylcytidine to regulate photosynthesis in rice. Cell Rep 2025; 44:115428. [PMID: 40085642 DOI: 10.1016/j.celrep.2025.115428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/16/2025] [Accepted: 02/21/2025] [Indexed: 03/16/2025] Open
Abstract
N4-acetylcytidine (ac4C) is a novel mRNA modification that enhances RNA stability and translation in mammals and plants. We previously identified ac4C in Arabidopsis, introduced by two homologs of human N-acetyltransferase 10 (NAT10). While ac4C influences leaf development in Arabidopsis, its role in rice is unclear. In this study, we identify OsNAT10 as the ac4C writer in rice. osnat10 mutants show developmental defects, including shorter roots, fewer tillers, and lower yield. Compared with wild type, ac4C-modified genes are less abundant in osnat10, particularly those related to photosynthesis. Additionally, osnat10 exhibits decreased photosynthetic capacity and reduced RNA stability and translation efficiency of ac4C target genes, like LIGHT-INDUCED RICE 1 (LIR1). Overexpressing OsLIR1 partially rescues osnat10 defects, underscoring OsNAT10's role in photosynthesis regulation. Our findings highlight ac4C's crucial function in photosynthesis and plant development, offering insights into epitranscriptomic modifications for crop improvement.
Collapse
Affiliation(s)
- Linjun Cai
- Hunan Key Laboratory of Plant Functional Genomics and Developmental Regulation, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Bin Li
- State Key Laboratory of Hybrid Rice, Hunan Hybrid Rice Research Center, Hunan Academy of Agricultural Sciences, Changsha, China
| | - Qiting Zhou
- Hunan Key Laboratory of Plant Functional Genomics and Developmental Regulation, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Juan Du
- Hunan Key Laboratory of Plant Functional Genomics and Developmental Regulation, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Wenxing Yang
- Hunan Key Laboratory of Plant Functional Genomics and Developmental Regulation, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Lisha Shen
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604, Singapore
| | - Chongsheng He
- Hunan Key Laboratory of Plant Functional Genomics and Developmental Regulation, College of Biology, Hunan University, Changsha, Hunan 410082, China.
| |
Collapse
|
126
|
Ji W, Xiong Y, Yang W, Shao Z, Guo X, Jin G, Su J, Zhou M. Transcriptomic profiling of blood platelets identifies a diagnostic signature for pancreatic cancer. Br J Cancer 2025:10.1038/s41416-025-02980-z. [PMID: 40133510 DOI: 10.1038/s41416-025-02980-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PaCa) is a deadly malignancy that is often diagnosed at an advanced stage, limiting treatment and reducing survival. There is an urgent need for convenient and accurate diagnostic markers for the early detection of PaCa. METHODS In this multicenter case-control study, we performed transcriptome analysis of 673 platelet samples from different in-house and public cohorts. RNA sequencing and RT-qPCR were used to discover and validate potential platelet biomarkers. A multi-gene signature was developed using binomial generalized linear model and independently validated in multicenter cohorts. RESULTS Two platelet RNAs, SCN1B and MAGOHB, consistently showed robust altered expression patterns between PaCa and healthy controls across cohorts, as confirmed by both RNA sequencing and RT-qPCR. The diagnostic two-RNA signature, PLA2Sig, demonstrated remarkable performance in detecting PaCa, with area under the receiver operating characteristic curve (AUC) values of 0.808, 0.900, 0.783, and 0.830 across multicenter cohorts. Furthermore, PLA2Sig effectively identified resectable stage I&II PaCa cases with an AUC of 0.812. Notably, PLA2Sig outperformed the traditional serum markers carcinoembryonic antigen and carbohydrate antigen 19-9 in distinguishing PaCa from healthy controls, and is complementary to established blood-based screening biomarkers. CONCLUSION These findings provide preliminary but promising evidence for the potential utility of platelet RNAs as an alternative non-invasive liquid biopsy tool for the early detection of PaCa.
Collapse
Affiliation(s)
- Weiping Ji
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yichun Xiong
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wei Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Zhuo Shao
- Department of General Surgery, Shanghai Changhai Hospital of Navy Medical University, Shanghai, 200438, China
| | - Xiaoling Guo
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Gang Jin
- Department of General Surgery, Shanghai Changhai Hospital of Navy Medical University, Shanghai, 200438, China
| | - Jianzhong Su
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Meng Zhou
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
127
|
Georges E, Ho W, Iturritza MU, Eory L, Malysz K, Sobhiafshar U, Archibald AL, Macqueen DJ, Shih B, Garrick D, Vernimmen D. Transcriptomic characterisation of acute myeloid leukemia cell lines bearing the same t(9;11) driver mutation reveals different molecular signatures. BMC Genomics 2025; 26:300. [PMID: 40133836 PMCID: PMC11938659 DOI: 10.1186/s12864-025-11415-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 02/28/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is the most common type of acute leukemia, accounting for 20% of cases in children and adolescents. Genome-wide studies have identified genes that are commonly mutated in AML, including many epigenetic regulators involved in either DNA methylation (DNMT3A, TET2, IDH1/2) or histone post-translational modifications (ASXL1, EZH2, MLL1). Several cell lines derived from AML patients are widely used in cancer research. Whether important differences in these cell lines exist remains poorly characterised. RESULTS Here, we used RNA sequencing (RNA-Seq) to contrast the transcriptome of four commonly used AML-derived cell lines: THP-1, NOMO-1, MOLM-13 bearing the common initiating t(9;11) translocation, and MV4.11 bearing the t(4;11) translocation. Gene set enrichment analyses and comparison of key transcription and epigenetic regulator genes revealed important differences in the transcriptome, distinguishing these AML models. Among these, we found striking differences in the expression of clusters of genes located on chromosome 19 encoding Zinc Finger (ZNF) transcriptional repressors. Low expression of many ZNF genes within these clusters is associated with poor survival in AML patients. CONCLUSION The present study offers a valuable resource by providing a detailed comparative characterisation of the transcriptome of cell lines within the same AML subtype used as models for leukemia research.
Collapse
Affiliation(s)
- Elise Georges
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - William Ho
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Miren Urrutia Iturritza
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Lel Eory
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Kamila Malysz
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Ulduz Sobhiafshar
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Alan L Archibald
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Daniel J Macqueen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Barbara Shih
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
- Present Address: Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| | - David Garrick
- INSERM UMR 1342, Institut de Recherche Saint Louis, Université Paris Cité, Paris, 75010, France
| | - Douglas Vernimmen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK.
| |
Collapse
|
128
|
Scofield S, Koshko L, Stilgenbauer L, Booms A, Berube R, Kassotis C, Lin CH, Jang H, Kim S, Stemmer P, Lempradl A, Sadagurski M. Integrative multi-omics analysis of metabolic dysregulation induced by occupational benzene exposure in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 971:179060. [PMID: 40068415 PMCID: PMC11928247 DOI: 10.1016/j.scitotenv.2025.179060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Type 2 Diabetes Mellitus (T2DM) is a significant public health burden. Emerging evidence links volatile organic compounds (VOCs), such as benzene to endocrine disruption and metabolic dysfunction. However, the effects of chronic environmentally relevant VOC exposures on metabolic health are still emerging. Building on our previous findings that benzene exposure at smoking levels (50 ppm) induces metabolic impairments in male mice, we investigated the effects of benzene exposure below OSHA's Occupational Exposure Limit (OEL) on metabolic health. Adult male C57BL/6 mice were exposed to 0.9 ppm benzene 8 h a day for 9 weeks. We assessed measures of metabolic homeostasis and conducted RNA and proteome sequencing on insulin-sensitive organs (liver, skeletal muscle, adipose tissue). At this dose, exposure caused significant metabolic disruptions, including hyperglycemia, hyperinsulinemia, and insulin resistance. Transcriptomic analysis of liver, muscle, and adipose tissue identified key changes in metabolic and immune pathways especially in liver. Proteomic analysis of the liver revealed mitochondrial dysfunction as a shared feature, with disruptions in oxidative phosphorylation, mitophagy, and immune activation. Comparative analysis with high-dose (50 ppm) exposure showed conserved and dose-specific transcriptomic changes in liver, particularly in metabolic and immune responses. Our study is the first to comprehensively assess the impacts of occupational benzene exposure on metabolic health, highlighting mitochondrial dysfunction as a central mechanism and the dose-dependent molecular pathways in insulin-sensitive organs driving benzene-induced metabolic imbalance. Our data indicate that the current OSHA OEL for benzene is insufficient and needs to be lowered, as they could result in adverse metabolic health in exposed workers, particularly men, following chronic exposure.
Collapse
Affiliation(s)
- Sydney Scofield
- Department of Biological Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, USA
| | - Lisa Koshko
- Department of Biological Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, USA
| | - Lukas Stilgenbauer
- Department of Biological Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, USA
| | - Alix Booms
- Van Andel Research Institute, Grand Rapids, MI, USA
| | - Roxanne Berube
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA; Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Christopher Kassotis
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA; Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Chung-Ho Lin
- School of Natural Resources, University of Missouri, Columbia, MO, USA
| | - Hyejeong Jang
- Department of Oncology, School of Medicine, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Seongho Kim
- Department of Oncology, School of Medicine, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Paul Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | | | - Marianna Sadagurski
- Department of Biological Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, USA; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
129
|
Sousa A, Besong OTO, Wachman EM, Taglauer ES, Beane JE, Kefella Y, Koo JS, Saia K, Jones HE, Zhang H. Placental transcriptome analysis in opioid-exposed versus non-opioid exposed pregnancies. Placenta 2025; 162:27-34. [PMID: 39983471 PMCID: PMC11908891 DOI: 10.1016/j.placenta.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/05/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
INTRODUCTION Opioid exposure during pregnancy may significantly alter gene expression in the placenta, potentially disrupting its function and influencing fetal brain development. These alterations may contribute to adverse outcomes such as neonatal opioid withdrawal syndrome (NOWS). In this study, we aim to systematically investigate the changes in placental gene expression associated with maternal opioid exposure to better understand the underlying molecular mechanisms and their implications for fetal health. METHODS Fresh placental tissue samples were collected from 18 opioid-exposed pregnancies and 26 non-opioid-exposed control pregnancies. Transcriptomic changes related to opioid exposure were assessed using RNA sequencing (RNA-seq). RESULTS Among the 16,172 genes detected, 55 showed differential expression (Padjusted < 0.25 or Punadjusted < 0.001) in opioid-exposed placentas. Gene Set Enrichment Analysis (GSEA) revealed that the differentially expressed genes were primarily associated with immune responses, neuronal development and function, as well as cell replication and division. Computational deconvolution using the PlacentaCellEnrich program identified significant enrichment of upregulated genes in decidual NK cells. Furthermore, integrative analysis of DNA methylation and gene expression showed an enrichment of differentially methylated genes among downregulated genes in opioid-exposed placentas. DISCUSSION Our findings suggest that opioid exposure during pregnancy may disrupt critical placental pathways, particularly those involved in immune responses. Future studies focusing on transcriptomic changes in specific placental cell types will be essential for fully understanding the structural and functional alterations in the placenta due to opioid exposure during pregnancy.
Collapse
Affiliation(s)
- Aneya Sousa
- Department of Pediatrics, Boston Medical Center, Boston, MA, USA
| | - Ojong Tabi Ojong Besong
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Elisha M Wachman
- Department of Pediatrics, Boston Medical Center, Boston, MA, USA.
| | | | - Jennifer E Beane
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Yohana Kefella
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Ji Sun Koo
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kelley Saia
- Department of Obstetrics and Gynecology, Boston Medical Center, Boston, MA, USA
| | - Hendree E Jones
- Department of Obstetrics & Gynecology, University of North Carolina, Chapel Hill, NC, USA
| | - Huiping Zhang
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
130
|
Nakajima-Koyama M, Kabata M, Lee J, Sogabe Y, Sakurai S, Hirota A, Kimura M, Nakamura T, Imoto Y, Kometani K, Hamazaki Y, Hiraoka Y, Saitou M, Nishida E, Yamamoto T. The balance between IFN-γ and ERK/MAPK signaling activities ensures lifelong maintenance of intestinal stem cells. Cell Rep 2025; 44:115286. [PMID: 39952238 DOI: 10.1016/j.celrep.2025.115286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 12/09/2024] [Accepted: 01/17/2025] [Indexed: 02/17/2025] Open
Abstract
While the intestinal epithelium has the highest cellular turnover rates in the mammalian body, it is also considered one of the tissues most resilient to aging-related disorders. Here, we reveal an innate protective mechanism that safeguards intestinal stem cells (ISCs) from environmental conditions in the aged intestine. Using in vivo phenotypic analysis, transcriptomics, and in vitro intestinal organoid studies, we show that age-dependent activation of interferon-γ (IFN-γ) signaling and inactivation of extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) signaling are responsible for establishing an equilibrium of Lgr5+ ISCs-between active and quiescent states-to preserve the ISC pool during aging. Furthermore, we show that differentiated cells have different sensitivities to each of the two signaling pathways, which may induce aging-related, functional, and metabolic changes in the body. Thus, our findings reveal an exquisitely balanced, age-dependent signaling mechanism that preserves stem cells at the expense of differentiated cells.
Collapse
Affiliation(s)
- May Nakajima-Koyama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa-Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan.
| | - Mio Kabata
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Joonseong Lee
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuko Sogabe
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Satoko Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akira Hirota
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa-Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Mizuki Kimura
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa-Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Tomonori Nakamura
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Hakubi Center for Advanced Research, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yusuke Imoto
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kohei Kometani
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoko Hamazaki
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Laboratory of Immunobiology, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuaki Hiraoka
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mitinori Saitou
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Eisuke Nishida
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa-Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan; RIKEN Center for Biosystems Dynamics Research (BDR), Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Medical-Risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto 606-8507, Japan.
| |
Collapse
|
131
|
Watanabe K, Yamano M, Miyamoto J, Ohue-Kitano R, Masujima Y, Sasahara D, Mouri Y, Kono N, Inuki S, Osakada F, Nagaoka K, Aoki J, Sugiura Y, Ohno H, Kondoh E, Kimura I. Maternal progesterone and adipose mPRε in pregnancy regulate the embryonic nutritional state. Cell Rep 2025; 44:115433. [PMID: 40085645 DOI: 10.1016/j.celrep.2025.115433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025] Open
Abstract
Sex steroid hormones such as progesterone play a pivotal role in reproductive functions and maintaining pregnancy; however, the impact of progesterone on the interaction between mother and embryo is unclear. Here, we demonstrate that the relationship between maternal progesterone and membrane progesterone receptor epsilon (mPRε) in adipose tissue regulates embryonic nutritional environment and growth after birth in mice. The activation of adipose mPRε by increased progesterone during pregnancy enhances maternal insulin resistance via prostaglandin production, efficiently providing glucose to embryos. Correspondingly, the offspring of mPRε-deficient mothers exhibited metabolic dysfunction, whereas mPRε-deficient mothers with high-fat diet-induced obesity exhibited improved insulin sensitivity. These findings establish the importance of progesterone as a nutritional regulator between mother and embryo. Additionally, mPRε may represent a modulator for treating pregnant glycemic control disorders such as gestational diabetes mellitus, as well as metabolic syndrome in offspring.
Collapse
Affiliation(s)
- Keita Watanabe
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mayu Yamano
- Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Junki Miyamoto
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo 183-8509, Japan
| | - Ryuji Ohue-Kitano
- Department of Biological & Environmental Chemistry, Kindai University, 11-6 Kayanomori, Iizuka City, Fukuoka 820-8555, Japan
| | - Yuki Masujima
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Daiki Sasahara
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Noster Inc., Kamiueno, Muko-shi, Kyoto 617-0006, Japan
| | - Yuki Mouri
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Nozomu Kono
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shunsuke Inuki
- Department of Bioorganic Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Fumitaka Osakada
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya 464-8601, Japan; Laboratory of Neural Information Processing, Institute for Advanced Research, Nagoya University, Nagoya 464-8601, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Kentaro Nagaoka
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan; Laboratory of Veterinary Physiology, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo 183-8509, Japan
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; Department of Moonshot Research and Development Program, Japan Science and Technology Agency, Chiyoda-ku, Tokyo 102-0076, Japan
| | - Yuki Sugiura
- Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroaki Ohno
- Department of Bioorganic Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Eiji Kondoh
- Laboratory of Neural Information Processing, Institute for Advanced Research, Nagoya University, Nagoya 464-8601, Japan; Department of Obstetrics and Gynecology, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-Ku, Kumamoto City, Kumamoto 860-8556, Japan
| | - Ikuo Kimura
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan; Department of Moonshot Research and Development Program, Japan Science and Technology Agency, Chiyoda-ku, Tokyo 102-0076, Japan.
| |
Collapse
|
132
|
Feng Q, Lin Z, Zhao D, Li M, Yang S, Xiang AP, Ye C, Yao C. Functional inhibition of core spliceosomal machinery activates intronic premature cleavage and polyadenylation of pre-mRNAs. Cell Rep 2025; 44:115376. [PMID: 40019833 DOI: 10.1016/j.celrep.2025.115376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/06/2025] [Accepted: 02/10/2025] [Indexed: 03/29/2025] Open
Abstract
The catalytic role of U6 snRNP in pre-mRNA splicing has been well established. In this study, we utilize an antisense morpholino oligonucleotide (AMO) specifically targeting catalytic sites of U6 snRNA to achieve functional knockdown of U6 snRNP in HeLa cells. The data show a significant increase in global intronic premature cleavage and polyadenylation (PCPA) events, similar to those observed with U1 AMO treatment, as demonstrated by mRNA 3'-seq analysis. Mechanistically, we provide evidence that U6 AMO-mediated splicing inhibition might be the driving force for PCPA as application of another specific AMO targeting U2 snRNP results in similar global PCPA effects. Together with our recently published findings that demonstrate the global inhibitory effect of U4 snRNP on intronic PCPA, our data highlight the critical role of splicing in suppressing intronic PCPA and support a model in which splicing and polyadenylation may compete with each other within introns during co-transcriptional mRNA processing.
Collapse
Affiliation(s)
- Qiumin Feng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Zejin Lin
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian 361102, China
| | - Danhui Zhao
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian 361102, China
| | - Mengzhao Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Sheng Yang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Congting Ye
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian 361102, China.
| | - Chengguo Yao
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China; Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Genetics and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
133
|
Hattori T, Taguchi K, Chaya R, Hamamoto S, Okada A, Yasui T. The role of osteopontin in modulating macrophage phagocytosis of calcium oxalate crystals. Urolithiasis 2025; 53:58. [PMID: 40131428 DOI: 10.1007/s00240-025-01732-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025]
Abstract
In inflammation, osteopontin (OPN) acts as both a stone matrix component for calcium oxalate (CaOx) crystal formation and an inflammatory mediator. While previous studies have demonstrated the individual roles of OPN and macrophages (Mφ) in renal CaOx stone formation during inflammation, their interaction remains poorly understood. This study aimed to elucidate the role of OPN in modulating Mφ function during crystal formation, using an ex vivo model. Bone marrow-derived macrophages (BMDM) were isolated from eight-week-old male C57BL/6J wild-type and OPN knockout mice. BMDMs from OPN-positive (BMDMOPN+) and OPN-negative (BMDMOPN-) mice were co-cultured with fluorescently labeled CaOx monohydrate (COM) crystals for phagocytosis assays and analyzed using the IN Cell Analyzer 6000. We further performed real-time quantitative reverse transcription PCR and RNA sequencing to identify gene expression profiles and clarify the role of OPN in Mφ function. The assay analysis demonstrated that phagocytosis rates were significantly higher in BMDMOPN- than in BMDMOPN+. Inflammatory markers, such as IL-6, TNF, CD44, were upregulated following COM exposure, and IL-6 expression was significantly lower in BMDMOPN- than in BMDMOPN+. RNA sequencing revealed that BMDMOPN- exhibited a less pro-inflammatory and more anti-inflammatory phenotype (Csf2low, Irf5low, Itgaxlow, Csf1high, Cd163high), resembling M2-like Mφs. Further functional analysis indicated that OPN knockdown in Mφs increased the S100 family and CREB signaling, which enhanced the M2-like phenotype shift and phagosome formation. In conclusion, OPN plays a critical role in enhancing pro-inflammatory Mφ function, potentially limiting COM phagocytosis. Modulating OPN expression in circulating Mφs may represent a therapeutic approach for kidney stone disease.
Collapse
Grants
- 23K27730, 23K08763, 23K08744, and 22H00486 Japan Society for the Promotion of Science
- 23K27730, 23K08763, 23K08744, and 22H00486 Japan Society for the Promotion of Science
- 23K27730, 23K08763, 23K08744, and 22H00486 Japan Society for the Promotion of Science
- 23K27730, 23K08763, 23K08744, and 22H00486 Japan Society for the Promotion of Science
- 23K27730, 23K08763, 23K08744, and 22H00486 Japan Society for the Promotion of Science
- 23K27730, 23K08763, 23K08744, and 22H00486 Japan Society for the Promotion of Science
Collapse
Affiliation(s)
- Tatsuya Hattori
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kazumi Taguchi
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | - Ryosuke Chaya
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Atsushi Okada
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahiro Yasui
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
134
|
Mendelsohn AI, Nikoobakht L, Bikoff JB, Costa RM. Segregated basal ganglia output pathways correspond to genetically divergent neuronal subclasses. Cell Rep 2025; 44:115454. [PMID: 40146776 DOI: 10.1016/j.celrep.2025.115454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
The basal ganglia control multiple sensorimotor behaviors through anatomically segregated and topographically organized subcircuits with outputs to specific downstream circuits. However, it is unclear how the anatomical organization of basal ganglia output circuits relates to the molecular diversity of cell types. Here, we demonstrate that the major output nucleus of the basal ganglia, the substantia nigra pars reticulata (SNr), is comprised of transcriptomically distinct subclasses that reflect its distinct progenitor lineages. We show that these subclasses are topographically organized within the SNr, project to distinct targets in the midbrain and hindbrain, and receive inputs from different striatal subregions. Finally, we show that these mouse subclasses are also identifiable in human SNr neurons, suggesting that the genetic organization of the SNr is evolutionarily conserved. These findings provide a unifying logic for how the developmental specification of diverse SNr neurons relates to the anatomical organization of basal ganglia circuits controlling specialized downstream brain regions.
Collapse
Affiliation(s)
- Alana I Mendelsohn
- Department of Neuroscience, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
| | - Laudan Nikoobakht
- Department of Neuroscience, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Jay B Bikoff
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rui M Costa
- Department of Neuroscience, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Allen Institute for Brain Science, Allen Institute, Seattle, WA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
135
|
Staples KJ, Ackland J, Lukose S, Angermann B, Belfield G, Belvisi M, Chaerkady R, Etal D, Heinson A, Hess S, Hristova VA, Hühn M, McCrae C, Muthas D, Öberg L, Ostridge K, Platt A, Spalluto CM, Watson A, Wilkinson T. Lung IL-13 gene signatures are associated with raised tissue eosinophils in COPD. Respir Res 2025; 26:114. [PMID: 40133927 PMCID: PMC11938775 DOI: 10.1186/s12931-025-03177-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/02/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The role of eosinophils in COPD and their utility as biomarkers for cytokine targeting monoclonal therapies remains unclear. We investigated the distribution of eosinophils across different tissue compartments in COPD and analysed gene expression to understand the possible mechanistic drivers of eosinophilic inflammation in COPD. METHODS Blood and BAL from ex-smoking volunteers with mild/moderate COPD (n = 31) and healthy ex-smoking controls (n = 20), and bronchial biopsy tissue in a subcohort (n = 19 and n = 8, respectively) was analysed. Differentially-expressed genes (DEGs) were characterised using RNASeq. Proteomic analysis of BAL was conducted using mass-spectrometry. RESULTS COPD subjects had more eosinophils in blood and lung tissue compared to controls, with increased eosinophil protein CLC/Galectin-10 in BAL. However, peripheral blood eosinophil counts related poorly to numbers in lung tissue (rho = -0.09192, p = 0.3541) or proportions in BAL (rho = 0.01762, p = 0.4632). Tissue IL-5Rα expression was higher in frequent exacerbators and related to tissue eosinophils, but not peripheral blood eosinophils. Higher blood eosinophils were associated with DEGs that differed with compartment. Higher tissue eosinophil levels were associated with IL-13-induced DEGs including POSTN in bronchial brushes and CCL26 in bronchial biopsies. Gene-set enrichment analysis on data from brushings revealed significant enrichment of IL-4/IL-13, but not IL-5, pathways associated with eosinophil presence. CONCLUSION Eosinophilic lung inflammation is related to exacerbation frequency, but lung eosinophils are not predicted by blood eosinophil counts in COPD. Our data suggest IL-13-mediated pathways may be responsible for the presence of tissue eosinophils in COPD. Further work to establish more predictive biomarkers of lung eosinophil biology are required to unlock this axis to optimised treatment.
Collapse
Affiliation(s)
- Karl J Staples
- Faculty of Medicine, University of Southampton, Southampton, UK.
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK.
| | - Jodie Ackland
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sruthymol Lukose
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Bastian Angermann
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Graham Belfield
- Translational Genomics, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Belvisi
- Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- NHLI, Imperial College, London, UK
| | - Raghothama Chaerkady
- Dynamic Omics, Centre of Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Damla Etal
- Translational Genomics, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ashley Heinson
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sonja Hess
- Dynamic Omics, Centre of Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Ventzislava A Hristova
- Dynamic Omics, Centre of Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Michael Hühn
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christopher McCrae
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Daniel Muthas
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lisa Öberg
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kristoffer Ostridge
- Faculty of Medicine, University of Southampton, Southampton, UK
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Adam Platt
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Alastair Watson
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Tom Wilkinson
- Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| |
Collapse
|
136
|
Fonseka O, Raja R, Ross C, Gare SR, Zhang J, Hille SS, King K, Ruiz-Velasco A, Kaur N, Chen X, Miller JM, Abouleisa RRE, Ou Q, Zou Z, Zhao X, Sotomayor-Flores C, Frank D, Swanton E, Pool MR, Missaglia S, Tavian D, Schiattarella GG, Wang T, Venetucci L, Pinali C, Rutter MK, Keavney BD, Cartwright EJ, Mohamed TMA, Müller OJ, Liu W. XBP1s-EDEM2 Prevents the Onset and Development of HFpEF by Ameliorating Cardiac Lipotoxicity. Circulation 2025. [PMID: 40130322 DOI: 10.1161/circulationaha.124.072194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/03/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Morbidity and mortality of heart failure with preserved ejection fraction (HFpEF) is increased in metabolic disorders. However, options for preventing and treating these prevalent outcomes are limited. Intramyocardial lipotoxicity contributes to cardiac dysfunction. Here, we investigate the mechanisms underlying endoplasmic reticulum degradation enhancing EDEM2 (endoplasmic reticulum degradation-enhancing alpha-mannosidase-like protein 2) regulation of cardiac lipid homeostasis and assess strategies that inhibit the incidence and progression of HFpEF. METHODS Metabolic stress was induced in C57BL/6 male mice using a high-fat diet and Nω-nitro-l-arginine methyl ester. The recombinant adeno-associated virus 9 delivery system was used for loss- and gain-of-function studies. Palmitic acid and oleic acid stimulation of rat cardiomyocytes and human induced pluripotent stem cell-derived cardiomyocytes imitated a condition of high lipids in vitro. Molecular mechanisms were investigated via RNA sequencing, mass spectrometry proteomics, lipidomic analyses, transmission electron microscopy, histology, and luciferase reporter assays. RESULTS In the human heart, we first detected lipid overload accompanied by a reduction of XBP1 (X-box binding protein 1) under metabolic stress. Thereafter, a decrease in EDEM2 was confirmed in human and mouse HFpEF hearts. Given that the spliced form of XBP1 (XBP1s) is a transcription factor, EDEM2 was identified as its new target in cardiomyocytes. EDEM2 knockdown mice manifested lipid droplet accumulation and higher levels of triglycerides and diglycerides in the myocardium, aggravating oxidative stress, hypertrophy, and the onset and progression of HFpEF under metabolic stress. XBP1s ablation mice displayed a similar myocardial lipid disturbance and cardiac phenotypes, which were reversed by EDEM2 overexpression. Mechanistically, the findings obtained from rat cardiomyocytes and human induced pluripotent stem cell-derived cardiomyocytes demonstrated that, in the presence of EDEM2, SEC23A mediated intracellular translocation of ATGL (adipose triglyceride lipase) under fatty acid stimulation, inhibiting ATGL degradation and excessive intracellular lipid droplets. Furthermore, the functional studies supported that EDEM2 prevention of lipid overload occurred in an ATGL-dependent manner. Therapeutically, cardiac XBP1s or EDEM2 restoration mitigated lipid deposition and preserved lipid profiles in the myocardium, thus preventing the development of HFpEF. CONCLUSIONS We demonstrate a cardioprotective mechanism regulating myocardial lipid homeostasis. The findings provide a promising therapeutic target to prevent and treat HfpEF, a condition with limited treatment options.
Collapse
Affiliation(s)
- Oveena Fonseka
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Rida Raja
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Claire Ross
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Sanskruti R Gare
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Jiayan Zhang
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Susanne S Hille
- Department of Internal Medicine, University of Kiel, Germany. V (S.S.H., D.F., O.J.M.)
- DZHK, German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany (S.S.H., O.J.M.)
| | - Katharine King
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Andrea Ruiz-Velasco
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Namrita Kaur
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Xinyi Chen
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Jessica M Miller
- Surgery Department, Baylor College of Medicine, Houston, TX (J.M.M., R.R.E.A., T.M.A.M.)
| | - Riham R E Abouleisa
- Surgery Department, Baylor College of Medicine, Houston, TX (J.M.M., R.R.E.A., T.M.A.M.)
| | - Qinghui Ou
- Institute of Molecular Cardiology, University of Louisville, KY (Q.O., T.M.A.M.)
| | - Zhiyong Zou
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Xiangjun Zhao
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Cristian Sotomayor-Flores
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (C.S.-F., G.G.S.)
- DZHK, German Centre for Cardiovascular Research, Partner Site Berlin, Germany (C.S.-F., G.G.S.)
| | - Derk Frank
- Department of Internal Medicine, University of Kiel, Germany. V (S.S.H., D.F., O.J.M.)
- Department of Internal Medicine III, University of Kiel, Germany. (D.F.)
| | - Eileithyia Swanton
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Martin R Pool
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Sara Missaglia
- Laboratory of Cellular Biochemistry and Molecular Biology, Università Cattolica del Sacro Cuore, Milan, Italy (S.M., D.T.)
| | - Daniela Tavian
- Laboratory of Cellular Biochemistry and Molecular Biology, Università Cattolica del Sacro Cuore, Milan, Italy (S.M., D.T.)
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (C.S.-F., G.G.S.)
- DZHK, German Centre for Cardiovascular Research, Partner Site Berlin, Germany (C.S.-F., G.G.S.)
- Translation Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (G.G.S.)
| | - Tao Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Luigi Venetucci
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Christian Pinali
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Martin K Rutter
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
- Diabetes, Endocrinology and Metabolism Centre, NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, UK. (M.K.R.)
| | - Bernard D Keavney
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
- Manchester Heart Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, UK. (B.D.K.)
| | - Elizabeth J Cartwright
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| | - Tamer M A Mohamed
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
- Surgery Department, Baylor College of Medicine, Houston, TX (J.M.M., R.R.E.A., T.M.A.M.)
- Institute of Molecular Cardiology, University of Louisville, KY (Q.O., T.M.A.M.)
| | - Oliver J Müller
- Department of Internal Medicine, University of Kiel, Germany. V (S.S.H., D.F., O.J.M.)
- DZHK, German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany (S.S.H., O.J.M.)
| | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, UK (O.F., R.R., C.R., S.R.G., J.Z., K.K., A.R.-V., N.K., X.C., Z.Z., X.Z., E.S., M.R.P., T.W., L.V., C.P., M.K.R., B.D.K., E.J.C., T.M.A.M., W.L.)
| |
Collapse
|
137
|
Cebrian-Silla A, Nascimento MA, Mancia W, Gonzalez-Granero S, Romero-Rodriguez R, Obernier K, Steffen DM, Lim DA, Garcia-Verdugo JM, Alvarez-Buylla A. Neural stem cell relay from B1 to B2 cells in the adult mouse ventricular-subventricular zone. Cell Rep 2025; 44:115264. [PMID: 40019835 DOI: 10.1016/j.celrep.2025.115264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/27/2024] [Accepted: 01/13/2025] [Indexed: 03/05/2025] Open
Abstract
Neurogenesis and gliogenesis continue in the ventricular-subventricular zone (V-SVZ) of the adult rodent brain. V-SVZ astroglial cells with apical contact with the ventricle (B1 cells) function as neural stem cells (NSCs). B1 cells sharply decline during early postnatal life; in contrast, neurogenesis decreases at a slower rate. Here, we show that a second population of astroglia (B2 cells) that do not contact the ventricle also function as NSCs in the adult mouse brain. B2 cell numbers increase postnatally, are sustained in adults, and decrease with aging. We reveal the transcriptomic profile of B1 and B2 cells and show that, like B1 cells, B2 cells can be quiescent or activated. Transplantation and lineage tracing of B2 cells demonstrate their function as primary progenitors for adult neurogenesis. This study reveals that NSC function is progressively relayed from B1 to B2 progenitors helping explain how neurogenesis is maintained into adult life.
Collapse
Affiliation(s)
- Arantxa Cebrian-Silla
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Marcos Assis Nascimento
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Walter Mancia
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Susana Gonzalez-Granero
- BTELab, Research Foundation of the General University Hospital of Valencia, Valencia 46014, Spain
| | - Ricardo Romero-Rodriguez
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kirsten Obernier
- Quantitative Biosciences Institute, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - David M Steffen
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Daniel A Lim
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jose Manuel Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, Paterna, 46980 Valencia, Spain
| | - Arturo Alvarez-Buylla
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
138
|
Tinkey RA, Smith BC, Habean ML, Williams JL. BATF2 is a regulator of interferon-γ signaling in astrocytes during neuroinflammation. Cell Rep 2025; 44:115393. [PMID: 40057949 DOI: 10.1016/j.celrep.2025.115393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/13/2025] [Accepted: 02/13/2025] [Indexed: 03/28/2025] Open
Abstract
Astrocytic interferon (IFN)γ signaling is associated with reduced neuroinflammation; however, downstream effectors responsible for regulating this protection are unknown. Here, we identify an IFN-specific transcription factor, basic leucine zipper ATF-like transcription factor (BATF)2, that plays a key role in modulating the consequences of IFNγ signaling in astrocytes. Chromatin immunoprecipitation sequencing revealed that BATF2 binds and prevents the overexpression of IFN regulatory factor (IRF)1 and IRF1 targets such as caspase-1. We also show that Batf2-/- mice exhibit exacerbated clinical disease severity in a murine model of central nervous system autoimmunity and express increased astrocyte-specific IRF1 and caspase-1, suggesting an amplified IFN response in vivo. Additionally, we demonstrate that BATF2 is expressed primarily in astrocytes within multiple sclerosis lesions and that this expression is colocalized with IRF1. Collectively, our results further support evidence of protective functions for IFNγ and implicate BATF2 as a key suppressor of overactive immune signaling in astrocytes during neuroinflammation.
Collapse
Affiliation(s)
- Rachel A Tinkey
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; School of Biomedical Sciences, Kent State University, Kent, OH 44240, USA
| | - Brandon C Smith
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Maria L Habean
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jessica L Williams
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
139
|
Outskouni Z, Prapa S, Goutas A, Klagkou E, Vatsellas G, Kosta A, Trachana V, Papathanasiou I. Comparative analysis of transcriptomic profiles of mesenchymal stem cells at the onset of senescence and after exposure to acute exogenous oxidative stress. Biochem Biophys Res Commun 2025; 754:151506. [PMID: 39999682 DOI: 10.1016/j.bbrc.2025.151506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/27/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
Cellular senescence can be triggered by a wide range of stress-inducing factors, including environmental and internal damaging events, such as oxidative stress. Moreover, stressed and senescent cells exhibit modifications in their transcriptional expression profile, but little is known regarding the common genes and pathways regulating these processes. Here, we analyzed the effects of long-term culture as well as exogenous acute oxidative stress on the transcriptional program of Wharton's jelly mesenchymal stem cells (WJ-MSCs). We demonstrate that, exposure to H2O2 compromised genomic stability and mitochondrial function in early passage WJ-MSCs, potentially initiating senescence to prevent cellular transformation. On the other hand, prolonged in vitro expansion of WJ-MSCs activated processes linked to integrins and extracellular matrix organization, possibly indicating the unfavorable consequences that senescence has on tissue integrity. Additionally, cells entering senescence and oxidative stressed young WJ-MSCs over-activated transcription factors related to permanent proliferative arrest and suppressed anti-senescence factors. Common differentially expressed genes in the late passage and H2O2-treated WJ-MSCs were implicated in DNA damage response and cell cycle arrest, which are known to trigger a senescent phenotype. Notably, the TP53INP1 gene emerged as a significantly upregulated gene in both late passage and H2O2-treated young WJ-MSCs, marking it as a potent senescence indicator. Silencing TP53INP1 mitigated the senescent phenotype, a role that appeared to be facilitated by autophagy regulation. Taken together, our results shed light on how transcriptomic changes govern MSCs' senescence program and identify key molecular drivers that could prove crucial for WJ-MSCs-based clinical applications.
Collapse
Affiliation(s)
- Zozo Outskouni
- Department of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, 41500, Greece
| | - Stavroula Prapa
- Department of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, 41500, Greece
| | - Andreas Goutas
- Department of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, 41500, Greece; Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Eleftheria Klagkou
- Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Giannis Vatsellas
- Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Artemis Kosta
- Microscopy Core Facility, Institut de Microbiologie de la Méditerranée (IMM), FR3479, CNRS, Aix-Marseille University, Marseille, France
| | - Varvara Trachana
- Department of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, 41500, Greece.
| | - Ioanna Papathanasiou
- Department of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, 41500, Greece.
| |
Collapse
|
140
|
O'Neill K, Shaw R, Bolger I, Tam OH, Phatnani H, Gale Hammell M. ALS molecular subtypes are a combination of cellular and pathological features learned by deep multiomics classifiers. Cell Rep 2025; 44:115402. [PMID: 40067829 DOI: 10.1016/j.celrep.2025.115402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/07/2025] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex syndrome with multiple genetic causes and wide variation in disease presentation. Despite this heterogeneity, large-scale genomics studies revealed that ALS postmortem samples can be grouped into a small number of subtypes, defined by transcriptomic signatures of mitochondrial dysfunction and oxidative stress (ALS-Ox), microglial activation and neuroinflammation (ALS-Glia), or TDP-43 pathology and associated transposable elements (ALS-TE). In this study, we present a deep ALS neural net classifier (DANCer) for ALS molecular subtypes. Applying DANCer to an expanded cohort from the NYGC ALS Consortium highlights two subtypes that strongly correlate with disease duration: ALS-TE in cortex and ALS-Glia in spinal cord. Finally, single-nucleus transcriptomes demonstrate that ALS subtypes are recapitulated in neurons and glia, with both ALS-wide and subtype-specific alterations in all cell types. In summary, ALS molecular subtypes represent a combination of cellular and pathological features that correlate with clinical features of ALS.
Collapse
Affiliation(s)
- Kathryn O'Neill
- Cold Spring Harbor Laboratory School of Biological Sciences, Cold Spring Harbor, NY 11724, USA
| | - Regina Shaw
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Neuroscience & Neuroscience Institute, NYU Langone Health, New York, NY 10016, USA
| | - Isobel Bolger
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Neuroscience & Neuroscience Institute, NYU Langone Health, New York, NY 10016, USA
| | - Oliver H Tam
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Neuroscience & Neuroscience Institute, NYU Langone Health, New York, NY 10016, USA.
| | - Hemali Phatnani
- New York Genome Center, New York, NY 10013, USA; Department of Neurology, Columbia University, New York, NY 10032, USA.
| | - Molly Gale Hammell
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Neuroscience & Neuroscience Institute, NYU Langone Health, New York, NY 10016, USA.
| |
Collapse
|
141
|
Dougherty MW, Hoffmann RM, Hernandez MC, Airan Y, Gharaibeh RZ, Herzon SB, Yang Y, Jobin C. Genome-scale CRISPR/Cas9 screening reveals the role of PSMD4 in colibactin-mediated cell cycle arrest. mSphere 2025; 10:e0069224. [PMID: 39918307 PMCID: PMC11934320 DOI: 10.1128/msphere.00692-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/14/2025] [Indexed: 03/26/2025] Open
Abstract
Colibactin is a genotoxic secondary metabolite produced by certain Enterobacteriaceae strains that populate the intestine and produces a specific mutational signature in human colonocytes. However, the host pathways involved in colibactin response remain unclear. To address this gap, we performed genome-wide CRISPR/Cas9 knockout screens and RNA sequencing utilizing live pks+ bacteria and a synthetic colibactin analog. We identified 20 enriched genes with a MAGeCK score of >2.0 in both screens, including proteasomal subunits (e.g., PSMG4 and PSMD4), RNA processing factors (e.g., SF1 and PRPF8), and RNA polymerase III (e.g., CRCP), and validated the role of PSMD4 in colibactin sensitization. PSMD4 knockout in HEK293T and HT-29 cells promoted cell viability and ameliorated G2-M cell cycle arrest but did not affect the amount of phosphorylated H2AX foci after exposure to synthetic colibactin 742. Consistent with these observations, PSMD4-/- cells had a significantly higher colony formation rate and bigger colony size than control cells after 742 exposure. These findings suggest that PSMD4 regulates cell cycle arrest following colibactin-induced DNA damage and that cells with PSMD4 deficiency may continue to replicate despite DNA damage, potentially increasing the risk of malignant transformation. IMPORTANCE Colibactin has been implicated as a causative agent of colorectal cancer. However, colibactin-producing bacteria are also present in many healthy individuals, leading to the hypothesis that some aspects of colibactin regulation or host response dictate the molecule's carcinogenic potential. Elucidating the host-response pathways involved in dictating cell fate after colibactin intoxication has been difficult, partially due to an inability to isolate the molecule. This study provides the first high-throughput CRISPR/Cas9 screening to identify genes conferring colibactin sensitivity. Here, we utilize both bacterial infection and a synthetic colibactin analog to identify genes directly involved in colibactin response. These findings provide insight into how differences in gene expression may render certain individuals more vulnerable to colibactin-initiated tumor formation after DNA damage.
Collapse
Affiliation(s)
- Michael W. Dougherty
- Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Ryan M. Hoffmann
- Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Maria C. Hernandez
- Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Yougant Airan
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
| | - Raad Z. Gharaibeh
- Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, USA
| | - Seth B. Herzon
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
- Departments of Pharmacology, Yale University, New Haven, Connecticut, USA
| | - Ye Yang
- Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Christian Jobin
- Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
- Department of Infectious Diseases and Immunology, University of Florida College of Medicine, Gainesville, Florida, USA
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
142
|
Petfalski E, Winz ML, Grelewska-Nowotko K, Turowski TW, Tollervey D. Multiple mechanisms of termination modulate the dynamics of RNAPI transcription. Cell Rep 2025; 44:115325. [PMID: 39999833 DOI: 10.1016/j.celrep.2025.115325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 11/01/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Transcription elongation is stochastic, driven by a Brownian ratchet, making it subject to changes in velocity. On the rDNA, multiple polymerases are linked by "torsional entrainment" generated by DNA rotation. We report that release of entrainment by co-transcriptional 3' end cleavage, is permissive for relative movement between polymerases, promoting pausing and backtracking. Subsequent termination (polymerase release) is facilitated by the 5' exonuclease Rat1 (Xrn2) and backtracked transcript cleavage by the RNA polymerase I (RNAPI) subunit Rpa12. These activities are reproduced in vitro. Short nascent transcripts close to the transcriptional start site, combined with nascent transcript folding energy, similarly facilitate RNAPI pausing. Nascent, backtracked transcripts at pause sites are terminated by forward and reverse "torpedoes": Rat1 and the exosome cofactor Trf4/5-Air1/2-Mtr4 polyadenylation (TRAMP), respectively. Topoisomerase 2 localizes adjacent to RNAPI pause sites, potentially allowing continued elongation by downstream polymerases. Mathematical modeling supported substantial premature termination. These basic insights into transcription in vivo will be relevant to many systems.
Collapse
Affiliation(s)
- Elisabeth Petfalski
- Centre for Cell Biology, School of Biological Sciences, The University of Edinburgh, Michael Swann Building, Edinburgh EH9 3BF, UK
| | - Marie-Luise Winz
- Centre for Cell Biology, School of Biological Sciences, The University of Edinburgh, Michael Swann Building, Edinburgh EH9 3BF, UK
| | | | - Tomasz W Turowski
- Institute of Biochemistry and Biophysics PAS, Pawińskiego 5A, 02-106 Warszawa, Poland.
| | - David Tollervey
- Centre for Cell Biology, School of Biological Sciences, The University of Edinburgh, Michael Swann Building, Edinburgh EH9 3BF, UK.
| |
Collapse
|
143
|
Puig-Barbe A, Dettmann S, Nirello VD, Moor H, Azami S, Edgar BA, Varga-Weisz P, Korzelius J, de Navascués J. A bHLH interaction code controls bipotential differentiation and self-renewal in the Drosophila gut. Cell Rep 2025; 44:115398. [PMID: 40089983 DOI: 10.1016/j.celrep.2025.115398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/04/2025] [Accepted: 02/14/2025] [Indexed: 03/18/2025] Open
Abstract
Multipotent adult stem cells balance self-renewal with differentiation into various cell types. How this balance is regulated at the transcriptional level is poorly understood. Here, we show that a network of basic helix-loop-helix (bHLH) transcription factors controls both stemness and bipotential differentiation in the Drosophila adult intestine. We find that homodimers of Daughterless (Da), a homolog of mammalian E proteins, maintain self-renewal of intestinal stem cells (ISCs), antagonizing the enteroendocrine fate promoted by heterodimers of Da and Scute (Sc; homolog of ASCL). The HLH factor Extramacrochaetae (Emc; homologous to Id proteins) promotes absorptive differentiation by titrating Da and Sc. Emc prevents the committed absorptive progenitor from dedifferentiating, underscoring the plasticity of these cells. Switching physical interaction partners in this way enables the active maintenance of stemness while priming stem cells for differentiation along two alternative fates. Such regulatory logic is likely operative in other bipotent stem cell systems.
Collapse
Affiliation(s)
- Aleix Puig-Barbe
- School of Biosciences, Cardiff University, The Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Svenja Dettmann
- DKFZ/ZMBH Alliance, University of Heidelberg, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; AbbVie Germany GmbH & Co. KG, 81 Mainzer Str., 65189 Wiesbaden, Frankfurt, Germany
| | - Vinícius Dias Nirello
- International Laboratory for Microbiome Host Epigenetics, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Helen Moor
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| | - Sina Azami
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Straße 9B, 50931 Köln, Germany
| | - Bruce A Edgar
- DKFZ/ZMBH Alliance, University of Heidelberg, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Huntsman Cancer Institute & Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Patrick Varga-Weisz
- International Laboratory for Microbiome Host Epigenetics, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil; School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| | - Jerome Korzelius
- DKFZ/ZMBH Alliance, University of Heidelberg, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Straße 9B, 50931 Köln, Germany; School of Biosciences, University of Kent, Canterbury CT2 7NZ, UK
| | - Joaquín de Navascués
- School of Biosciences, Cardiff University, The Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK; School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK.
| |
Collapse
|
144
|
Wan Q, Yao R, Zhao Y, Xu L. JA and ABA signaling pathways converge to protect plant regeneration in stress conditions. Cell Rep 2025; 44:115423. [PMID: 40088448 DOI: 10.1016/j.celrep.2025.115423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 01/02/2025] [Accepted: 02/20/2025] [Indexed: 03/17/2025] Open
Abstract
In cuttings, detached leaves or stems are exposed to many stresses during the root regeneration process. Here, we show that the detached Arabidopsis thaliana leaf can tolerate mild osmotic stress and still regenerate roots. Under stress conditions, wounding and stress upregulate the jasmonate (JA) signaling pathway transcription factor gene MYC2 and the abscisic acid (ABA) signaling pathway transcription factor gene ABA INSENSITIVE5 (ABI5). The MYC2-ABI5 complex upregulates the expression of β-GLUCOSIDASE18 (BGLU18), which releases ABA from ABA glucose ester, resulting in ABA accumulation in the detached leaf. Mutations in MYC2, ABI5, and BGLU18 lead to the loss of stress tolerance and defects in root regeneration under osmotic stress. The successive application of JA and ABA can enhance the root regeneration ability in Arabidopsis and poplar cuttings. Overall, the JA-mediated wound signaling pathway and the ABA-mediated stress signaling pathway collaboratively amplify ABA signals to protect root regeneration under stress conditions.
Collapse
Affiliation(s)
- Qihui Wan
- Key Laboratory of Plant Carbon Capture, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, 300 Fenglin Road, Shanghai 200032, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Ruifeng Yao
- State Key Laboratory of Chemo and Biosensing, Hunan Provincial Key Laboratory of Plant Functional Genomics and Developmental Regulation, Hunan Research Center of the Basic Discipline for Cell Signaling, College of Biology, Hunan University, Changsha 410082, China
| | - Yang Zhao
- Key Laboratory of Plant Carbon Capture, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, 300 Fenglin Road, Shanghai 200032, China; Shanghai Center for Plant Stress Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Lin Xu
- Key Laboratory of Plant Carbon Capture, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, 300 Fenglin Road, Shanghai 200032, China.
| |
Collapse
|
145
|
Chang J, Campbell-Hanson KR, Vanneste M, Bartschat NI, Nagel R, Arnadottir AK, Vu HN, Montgomery C, Yevdash J, Jiang J, Bhinu A, Helverson A, Henry MD, Steingrímsson E, Weigel RJ, Cornell RA, Kenny C. Antagonistic roles for MITF and TFE3 in melanoma plasticity. Cell Rep 2025; 44:115474. [PMID: 40138313 DOI: 10.1016/j.celrep.2025.115474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 01/06/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Melanoma cells can switch from a melanocytic and proliferative state to a mesenchymal and invasive state and back again. This plasticity drives intratumoral heterogeneity, progression, and therapeutic resistance. Microphthalmia-associated transcription factor (MITF) promotes the melanocytic/proliferative phenotype, but factors that drive the mesenchymal/invasive phenotype and the mechanisms that effect the switch between cell states are unclear. Here, we identify the MITF paralog, TFE3, and the non-canonical mTORC1 pathway as regulators of the mesenchymal state. We show that TFE3 expression drives the metastatic phenotype in melanoma cell lines and tumors. Deletion of TFE3 in MITF-low melanoma cell lines suppresses their ability to migrate and metastasize. Further, MITF suppresses the mesenchymal phenotype by directly or indirectly activating expression of FNIP1, FNIP2, and FLCN, which encode components of the non-canonical mTORC1 pathway, thereby promoting cytoplasmic retention and lysosome-mediated degradation of TFE3. These findings highlight a molecular pathway controlling melanoma plasticity and invasiveness.
Collapse
Affiliation(s)
- Jeremy Chang
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | - Marion Vanneste
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nicholas I Bartschat
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ryan Nagel
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Asdis K Arnadottir
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Hong Nhung Vu
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Collin Montgomery
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Julius Yevdash
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Jiarui Jiang
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ardith Bhinu
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Annika Helverson
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Michael D Henry
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Eiríkur Steingrímsson
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ronald J Weigel
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Robert A Cornell
- Department of Oral Health Sciences, University of Washington, School of Dentistry, Seattle, WA, USA
| | - Colin Kenny
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
146
|
Xie K, Wang C, Scifo E, Pearson B, Ryan D, Henzel K, Markert A, Schaaf K, Mi X, Tian X, Jia J, Wang M, Bonn S, Schölling M, Möhl C, Bano D, Zhou Y, Ehninger D. Intermittent fasting boosts sexual behavior by limiting the central availability of tryptophan and serotonin. Cell Metab 2025:S1550-4131(25)00104-4. [PMID: 40157367 DOI: 10.1016/j.cmet.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 12/02/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
Abstract
Aging affects reproductive capabilities in males through physiological and behavioral alterations, including endocrine changes and decreased libido. In this study, we investigated the influence of intermittent fasting (IF) on these aging-related declines, using male C57BL/6J mice. Our findings revealed that IF significantly preserved reproductive success in aged mice, not by improving traditional reproductive metrics such as sperm quality or endocrine functions but by enhancing mating behavior. This behavioral improvement was attributed to IF's ability to counter age-dependent increases in serotonergic inhibition, primarily through the decreased supply of the serotonin precursor tryptophan from the periphery to the brain. Our research underscores the potential of dietary interventions like IF in mitigating age-associated declines in male reproductive health and suggests a novel approach to managing conditions related to reduced sexual desire, highlighting the complex interplay between diet, metabolism, and reproductive behavior.
Collapse
Affiliation(s)
- Kan Xie
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Chengfeng Wang
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China
| | - Enzo Scifo
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Brandon Pearson
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Devon Ryan
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Kristin Henzel
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Astrid Markert
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Kristina Schaaf
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Xue Mi
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China
| | - Xin Tian
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China
| | - Jiajia Jia
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China
| | - Meiqin Wang
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China
| | - Stefan Bonn
- Institute of Medical Systems Biology, Hamburg Center for Biomedical AI (bAIome), Molecular Neurobiology Hamburg (ZMNH), and Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Manuel Schölling
- Image and Data Analysis Facility, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Christoph Möhl
- Image and Data Analysis Facility, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Daniele Bano
- Aging and Neurodegeneration Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Yu Zhou
- Department of Intensive Care Unit, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, Shandong, China; Institute of Brain Sciences and Related Disorders, Medical College of Qingdao University, Qingdao 266071, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, Shandong, China.
| | - Dan Ehninger
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany.
| |
Collapse
|
147
|
Kane MA, Birmingham KG, Yeoman B, Patel N, Sperinde H, Molley TG, Beri P, Tuler J, Kumar A, Klein S, Zare S, Wallace A, Katira P, Engler AJ. Adhesion strength of tumor cells predicts metastatic disease in vivo. Cell Rep 2025; 44:115359. [PMID: 40049163 DOI: 10.1016/j.celrep.2025.115359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/26/2024] [Accepted: 02/06/2025] [Indexed: 03/29/2025] Open
Abstract
Although only a fraction of tumor cells contribute to metastatic disease, no prognostic biomarkers currently exist to identify these cells. We show that a physical marker-adhesion strength-predicts metastatic potential in a mouse breast cancer model and that it may stratify human disease. Cells disseminating from murine mammary tumors are weakly adherent, and, when pre-sorted by adhesion, primary tumors created from strongly adherent cells exhibit fewer lung metastases than weakly adherent cells do. We demonstrate that admixed cancer lines can be separated by label-free adhesive signatures. When applied to murine metastatic tumors, adhesion retrospectively predicts metastatic disease with 100% specificity, 85% sensitivity, and area under the curve (AUC) of 0.94. Cells from human reduction mammoplasties have a higher adhesion strength versus resected human tumors, which may also be stratified between invasive and more indolent cancers. Thus, highly metastatic cells may have a distinct physical phenotype that may be a predictive marker of clinical outcomes.
Collapse
Affiliation(s)
- Madison A Kane
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | | | - Benjamin Yeoman
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Department of Mechanical Engineering, San Diego State University, San Diego, CA 92182, USA
| | - Neal Patel
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
| | - Hayley Sperinde
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Thomas G Molley
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Pranjali Beri
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
| | - Jeremy Tuler
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Aditya Kumar
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
| | - Sarah Klein
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
| | - Somaye Zare
- Department of Pathology, UC San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, UC San Diego, La Jolla, CA 92093, USA
| | - Anne Wallace
- Department of Surgery, UC San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, UC San Diego, La Jolla, CA 92093, USA
| | - Parag Katira
- Department of Mechanical Engineering, San Diego State University, San Diego, CA 92182, USA; Computational Science Research Center, San Diego State University, San Diego, CA 92182, USA
| | - Adam J Engler
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA; Department of Pathology, UC San Diego, La Jolla, CA 92093, USA; Department of Surgery, UC San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, UC San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.
| |
Collapse
|
148
|
Lisicka W, Earley ZM, Sifakis JJ, Erickson SA, Mattingly JR, Wu-Woods NJ, Krishnamurthy SR, Belkaid Y, Ismagilov RF, Cyster JG, Riesenfeld SJ, Bendelac A, Jabri B. Immunoglobulin A controls intestinal virus colonization to preserve immune homeostasis. Cell Host Microbe 2025:S1931-3128(25)00087-3. [PMID: 40154490 DOI: 10.1016/j.chom.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/26/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
Abstract
Immunoglobulin A (IgA) is the predominant immunoglobulin isotype in mammals, primarily secreted at type I mucosal surfaces. Despite its abundance, the precise role of secretory IgA in the intestinal lumen, where it coats a diverse array of commensal microbiota, has remained elusive. Our study reveals that germinal center IgA responses are essential for preventing chronic colonization of the gut by specific viruses. In the absence of IgA, chronic viral colonization triggers an antigen-driven expansion of CD8αβ+ intraepithelial lymphocytes (IELs). Although these IELs are unable to clear the virus, they contribute to maintaining homeostasis by regulating its load and type I interferon responses. Consequently, IgA deficiency increases susceptibility to colitis in genetically susceptible hosts or following chemical induction but only in the presence of viral pathobionts requiring IgA for their clearance. These findings underscore the potential vulnerability of IgA-deficient individuals to immunopathology when exposed to selective viral pathobionts.
Collapse
Affiliation(s)
- Wioletta Lisicka
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Zachary M Earley
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph J Sifakis
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Steven A Erickson
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Jonathan R Mattingly
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Natalie J Wu-Woods
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Siddharth R Krishnamurthy
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rustem F Ismagilov
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jason G Cyster
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Samantha J Riesenfeld
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA; Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Albert Bendelac
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Bana Jabri
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA; Department of Pathology, University of Chicago, Chicago, IL, USA; Paris City University, Imagine Institute, Paris, France.
| |
Collapse
|
149
|
Chen P, Lin L, Lin X, Liao K, Qiang J, Wang Z, Wu J, Li Y, Yang L, Yao N, Song H, Hong Y, Liu WH, Zhang Y, Chang X, Du D, Xiao C. A Csde1-Strap complex regulates plasma cell differentiation by coupling mRNA translation and decay. Nat Commun 2025; 16:2906. [PMID: 40133358 PMCID: PMC11937441 DOI: 10.1038/s41467-025-58212-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Upon encountering antigens, B cells may undergo multiple differentiation paths, including becoming plasma cells and memory B cells. Although it is well-known that transcription factors govern gene expression programs underpinning these fate decisions in transcriptional level, the role of post-transcriptional regulators, with a focus on RNA-binding proteins, in the fate determination are lesser known. Here we find by RNA interactome capture-coupled CRISPR/Cas9 functional screening that the Csde1-Strap complex plays an important role in plasma cell differentiation. Mechanistically, the Csde1-Strap complex establishes the expression kinetics of Bach2, a key regulator of plasma cell differentiation. Bach2 expression is rapidly induced to promote B cell expansion and then decreased to initiate plasma cell differentiation. The Csde1-Strap interaction is critical for their binding to Bach2 mRNA to couple its decay with translation to restrain the magnitude and duration of Bach2 protein expression. In the absence of Csde1 or Strap, Bach2 translation is de-coupled from mRNA decay, leading to elevated and prolonged expression of Bach2 protein and impaired plasma cell differentiation. This study thus establishes the functional RBP landscape in B cells and illustrates the fundamental importance of controlling protein expression kinetics in cell fate determination.
Collapse
Affiliation(s)
- Pengda Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Lianghua Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xinyong Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Kunyu Liao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jiali Qiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zhizhang Wang
- Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yang Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Liang Yang
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
| | - Nan Yao
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
| | - Huilin Song
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yazhen Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Wen-Hsien Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
| | - Yaoyang Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| | - Xing Chang
- Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| | - Dan Du
- State Key Laboratory of Cellular Stress Biology, Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
| | - Changchun Xiao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
150
|
Lee EJ, Kim M, Park S, Shim JH, Cho HJ, Park JA, Park K, Lee D, Kim JH, Jeong H, Matsuzaki F, Kim SY, Kim J, Yang H, Lee JS, Kim JW. Restoration of retinal regenerative potential of Müller glia by disrupting intercellular Prox1 transfer. Nat Commun 2025; 16:2928. [PMID: 40133314 PMCID: PMC11937340 DOI: 10.1038/s41467-025-58290-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Individuals with retinal degenerative diseases struggle to restore vision due to the inability to regenerate retinal cells. Unlike cold-blooded vertebrates, mammals lack Müller glia (MG)-mediated retinal regeneration, indicating the limited regenerative capacity of mammalian MG. Here, we identify prospero-related homeobox 1 (Prox1) as a key factor restricting this process. Prox1 accumulates in MG of degenerating human and mouse retinas but not in regenerating zebrafish. In mice, Prox1 in MG originates from neighboring retinal neurons via intercellular transfer. Blocking this transfer enables MG reprogramming into retinal progenitor cells in injured mouse retinas. Moreover, adeno-associated viral delivery of an anti-Prox1 antibody, which sequesters extracellular Prox1, promotes retinal neuron regeneration and delays vision loss in a retinitis pigmentosa model. These findings establish Prox1 as a barrier to MG-mediated regeneration and highlight anti-Prox1 therapy as a promising strategy for restoring retinal regeneration in mammals.
Collapse
Affiliation(s)
- Eun Jung Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- Celliaz Ltd., Daejeon, South Korea
| | - Museong Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Sooyeon Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- Celliaz Ltd., Daejeon, South Korea
| | | | - Hyun-Ju Cho
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
- KRIBB School, University of Science and Technology, Daejeon, South Korea
| | | | - Kihyun Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Dongeun Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Jeong Hwan Kim
- Korea Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Haeun Jeong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Fumio Matsuzaki
- Laboratory for Cell Asymmetry, RIKEN Centre for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Department of Aging Science and Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Seon-Young Kim
- Korea Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Jaehoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Hanseul Yang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Jeong-Soo Lee
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
- KRIBB School, University of Science and Technology, Daejeon, South Korea
| | - Jin Woo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.
- Celliaz Ltd., Daejeon, South Korea.
| |
Collapse
|