1
|
Lo Presti E, Cupaioli F, Scimeca D, Unti E, Di Martino V, Daidone R, Amata M, Scibetta N, Soucie E, Meraviglia S, Iovanna J, Dusetti N, De Gaetano A, Merelli I, Di Mitri R. The pancreatic tumor microenvironment of treatment-naïve patients causes a functional shift in γδ T cells, impairing their anti-tumoral defense. Oncoimmunology 2025; 14:2466301. [PMID: 39945298 PMCID: PMC11834455 DOI: 10.1080/2162402x.2025.2466301] [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: 07/24/2024] [Revised: 01/14/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents a unique challenge for researchers due to its late diagnosis caused by vague symptoms and lack of early detection markers. Additionally, PDAC is characterized by an immunosuppressive microenvironment (TME), making it a difficult tumor to treat. While γδ T cells have shown potential for anti-tumor activity, conflicting studies exist regarding their effectiveness in pancreatic cancer. This study aims to explore the hypothesis that the PDAC TME hinders the anti-tumor capabilities of γδ T cells through blockade of cytotoxic functions. For this reason, we chose to enroll PDAC treatment-naive patients to avoid the possibility of therapy modifying the TME. By flow cytometry, our research findings indicate that the presence of γδ T cells among CD45+ cells in tumor tissue is lower compared to CD66+ cells, but higher than in blood. Circulating Vδ1 T cells exhibit a terminal effector memory phenotype (TEMRA) more than Vδ2 T cells. Interestingly, Vδ1 and Vδ2 T cells appear to be more prevalent at different stages of tumor development. In our in vitro culture using conditioned medium derived from Patient-derived organoids ;(PDOs), we observed a shift in expression markers in γδ T cells of healthy individuals toward an activation and exhaustion phenotype, as confirmed by scRNA-seq analysis extracted from a public database. A deeper understanding of γδ T cells in PDAC could be valuable for developing novel therapies aimed at mitigating the impact of the pancreatic tumor microenvironment on this cell population.
Collapse
Affiliation(s)
- Elena Lo Presti
- National Research Council of Italy (CNR), Institute for Biomedical Research and Innovation (IRIB), Palermo, Italy
| | - Francesca Cupaioli
- National Research Council of Italy, Bioinformatics Research Unit, Institute for Biomedical Technologies Segrate, Milan, Italy
| | - Daniela Scimeca
- Gastroenterology and Endoscopy Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Elettra Unti
- ‘Anatomic-pathology Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Vincenzo Di Martino
- Immunohaematology and Transfusion Medicine Unit, Imperia Hospital ASL1 Imperiese, Imperia, Italy
| | - Rossella Daidone
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Michele Amata
- Gastroenterology and Endoscopy Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Nunzia Scibetta
- ‘Anatomic-pathology Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Erinn Soucie
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Serena Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy
| | - Juan Iovanna
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Nelson Dusetti
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Andrea De Gaetano
- National Research Council of Italy (CNR), Institute for Biomedical Research and Innovation (IRIB), Palermo, Italy
- National Research Council of Italy, Institute for Systems Analysis and Computer Science “A. Ruberti, ” BioMatLab (Biomathematics Laboratory), Rome, Italy
- Department of Mathematics, Mahidol University, Bangkok, Thailand
| | - Ivan Merelli
- National Research Council of Italy, Bioinformatics Research Unit, Institute for Biomedical Technologies Segrate, Milan, Italy
| | - Roberto Di Mitri
- Gastroenterology and Endoscopy Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| |
Collapse
|
2
|
Muñoz Sandoval D, Bach FA, Ivens A, Harding AC, Smith NL, Mazurczyk M, Themistocleous Y, Edwards NJ, Silk SE, Barrett JR, Cowan GJ, Napolitani G, Savill NJ, Draper SJ, Minassian AM, Nahrendorf W, Spence PJ. Plasmodium falciparum infection induces T cell tolerance that is associated with decreased disease severity upon re-infection. J Exp Med 2025; 222:e20241667. [PMID: 40214640 PMCID: PMC11987708 DOI: 10.1084/jem.20241667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/18/2024] [Accepted: 03/12/2025] [Indexed: 04/14/2025] Open
Abstract
Immunity to severe malaria is acquired quickly, operates independently of pathogen load, and represents a highly effective form of disease tolerance. The mechanism that underpins tolerance remains unknown. We used a human rechallenge model of falciparum malaria in which healthy adult volunteers were infected three times over a 12 mo period to track the development of disease tolerance in real-time. We found that parasitemia triggered a hardwired innate immune response that led to systemic inflammation, pyrexia, and hallmark symptoms of clinical malaria across the first three infections of life. In contrast, a single infection was sufficient to reprogram T cell activation and reduce the number and diversity of effector cells upon rechallenge. Crucially, this did not silence stem-like memory cells but instead prevented the generation of cytotoxic effectors associated with autoinflammatory disease. Tolerized hosts were thus able to prevent collateral tissue damage in the absence of antiparasite immunity.
Collapse
Affiliation(s)
- Diana Muñoz Sandoval
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
- Instituto de Microbiologia, Universidad San Francisco de Quito, Quito, Ecuador
| | - Florian A. Bach
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Alasdair Ivens
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Adam C. Harding
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Natasha L. Smith
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Michalina Mazurczyk
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | | | - Sarah E. Silk
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Jordan R. Barrett
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Graeme J.M. Cowan
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Giorgio Napolitani
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nicholas J. Savill
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Angela M. Minassian
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Wiebke Nahrendorf
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Philip J. Spence
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
3
|
Fan X, Li H. Integration of Single-Cell and Spatial Transcriptomic Data Reveals Spatial Architecture and Potential Biomarkers in Alzheimer's Disease. Mol Neurobiol 2025; 62:5395-5412. [PMID: 39543008 DOI: 10.1007/s12035-024-04617-3] [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: 05/21/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by the gradual loss of neurons and the accumulation of amyloid plaques and neurofibrillary tangles. Despite advancements in the understanding of AD's pathophysiology, the cellular organization and interactions in the prefrontal cortex (PFC) remain elusive. Eight single-cell RNA sequencing (scRNA-seq) datasets from both normal controls and individuals with AD were harmonized. Stringent preprocessing protocols were implemented to uphold dataset integrity. Unsupervised clustering and annotation revealed 22 distinct cell clusters corresponding to 19 unique cell types. The spatial architecture of the PFC region was constructed using the CARD tool. Further analyses encompassed trajectory examination of Oligodendrocyte subtypes, evaluation of regulon activity scores, and spot clustering within white matter regions (WM). Differential expression analysis and functional enrichment assays unveiled molecular signatures linked to AD progression and were validated using microarray data sourced from neurodegenerative disorder patients. Our investigation employs scRNA-seq and spatial transcriptomics to uncover the cellular atlas and spatial architecture of the human PFC in AD. Moreover, our results indicate that Oligodendrocytes are more prevalent in AD patients, showcasing diverse subtypes and spatial organization within WM regions. Each subtype appears to be associated with distinct biological processes and transcriptional regulators, shedding light on their involvement in AD pathology. Notably, the Oligodendrocyte_C6 subtype is linked to neurological damage in AD patients, characterized by heightened expression of genes involved in cell-cell connections, cell membrane stability, and myelination. Additionally, 12 target genes regulated by NFIA were identified, which are upregulated in AD patients and associated with disease progression. Elevated PLXDC2 expression in peripheral blood was also identified, suggesting its potential as a non-invasive biomarker for early AD detection. Our study provides novel insights into the role of Oligodendrocytes in AD and highlights the potential of PLXDC2 as a blood biomarker for non-invasive diagnosis and monitoring of AD patients.
Collapse
Affiliation(s)
- Xing Fan
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong, 226001, PR, China
| | - Huamei Li
- Department of Rheumatology and Immunology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, PR, China.
| |
Collapse
|
4
|
Joshi T, Chan YO, Qiao Z, Kheirandish-Gozal L, Gozal D, Khalyfa A. Circulating exosomes in pediatric obstructive sleep apnea with or without neurocognitive deficits and their effects on a 3D-blood-brain barrier spheroid model. Exp Neurol 2025; 387:115188. [PMID: 39986553 DOI: 10.1016/j.expneurol.2025.115188] [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: 10/19/2024] [Revised: 01/31/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
Obstructive sleep apnea (OSA) in children is linked to cognitive impairments, potentially due to blood-brain barrier (BBB) dysfunction. Exosomes, small vesicles released by most cells, reflect cellular changes. This study examined the effects of exosomes from children with OSA, with or without cognitive deficits, on neurovascular unit (NVU) models. Twenty-six children were categorized into three groups: healthy controls (Cont, n = 6), OSA without cognitive deficits (OSA-NG, n = 10), and OSA with neurocognitive deficits (OSA-POS, n = 10). Plasma exosomes were characterized and applied to human 3D NVU spheroids for 24 h. Barrier integrity, permeability, and angiogenesis were assessed using trans-endothelial electrical resistance (TEER), tight junction integrity, and tube formation assays. Single-nucleus RNA sequencing (snRNA-seq) and bioinformatics, including CellChat analysis, identified intercellular signaling pathways. Results showed that exosomes from OSA-POS children disrupted TEER, increased permeability, and impaired ZO1 staining in spheroids, compared to the other groups. Both OSA-POS and OSA-NG exosomes increased permeability in NVU cells in monolayer and microfluidic BBB models. snRNA-seq analysis further revealed distinct cell clusters and pathways associated with the different groups. This 3D NVU spheroid model provides a robust platform to study BBB properties and the role of exosomes in OSA. These findings suggest that integrating snRNA-seq with exosome studies can uncover mechanisms underlying neurocognitive dysfunction in pediatric OSA, potentially leading to personalized therapeutic approaches.
Collapse
Affiliation(s)
- Trupti Joshi
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA; Christophers S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
| | - Yen On Chan
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA; Christophers S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
| | - Zhuanhong Qiao
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
| | | | - David Gozal
- Department of Pediatrics, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| | - Abdelnaby Khalyfa
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States of America.
| |
Collapse
|
5
|
Kim HY, Rosenthal SB, Liu X, Miciano C, Hou X, Miller M, Buchanan J, Poirion OB, Chilin-Fuentes D, Han C, Housseini M, Carvalho-Gontijo Weber R, Sakane S, Lee W, Zhao H, Diggle K, Preissl S, Glass CK, Ren B, Wang A, Brenner DA, Kisseleva T. Multi-modal analysis of human hepatic stellate cells identifies novel therapeutic targets for metabolic dysfunction-associated steatotic liver disease. J Hepatol 2025; 82:882-897. [PMID: 39522884 DOI: 10.1016/j.jhep.2024.10.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/09/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatotic liver disease ranges from metabolic dysfunction-associated steatotic liver (MASL) to metabolic dysfunction-associated steatohepatitis (MASH) with fibrosis. Transdifferentiation of hepatic stellate cells (HSCs) into fibrogenic myofibroblasts plays a critical role in the pathogenesis of MASH liver fibrosis. We compared transcriptome and chromatin accessibility of human HSCs from NORMAL, MASL, and MASH livers at single-cell resolution. We aimed to identify genes that are upregulated in activated HSCs and to determine which of these genes are key in the pathogenesis of MASH fibrosis. METHODS Eighteen human livers were profiled using single-nucleus (sn)RNA-seq and snATAC-seq. High priority targets were identified, then tested in 2D human HSC cultures, 3D human liver spheroids, and HSC-specific gene knockout mice. RESULTS MASH-enriched activated HSC subclusters are the major source of extracellular matrix proteins. We identified a set of concurrently upregulated and more accessible core genes (GAS7, SPON1, SERPINE1, LTBP2, KLF9, EFEMP1) that drive activation of HSCs. Expression of these genes was regulated via crosstalk between lineage-specific (JUNB/AP1), cluster-specific (RUNX1/2) and signal-specific (FOXA1/2) transcription factors. The pathological relevance of the selected targets, such as SERPINE1 (PAI-1), was demonstrated using dsiRNA-based HSC-specific gene knockdown or pharmacological inhibition of PAI-1 in 3D human MASH liver spheroids, and HSC-specific Serpine1 knockout mice. CONCLUSION This study identified novel gene targets and regulatory mechanisms underlying activation of fibrogenic HSCs in MASH, and demonstrated that genetic or pharmacological inhibition of select genes suppressed liver fibrosis. IMPACT AND IMPLICATIONS Herein, we present the results of a multi-modal sequencing analysis of human hepatic stellate cells (HSCs) from NORMAL, MASL (metabolic dysfunction-associated steatotic liver), and metabolic dysfunction-associated steatohepatitis (MASH) livers. We identified additional subclusters that were not detected by previous studies and characterized the mechanism by which HSCs are activated in MASH livers, including the transcriptional machinery that induces the transdifferentiation of HSCs into myofibroblasts. For the first time, we described the pathogenic role of activated HSC-derived PAI-1 (a product of the SERPINE1 gene) in the development of MASH liver fibrosis. Targeting the RUNX1/2-SERPINE1 axis could be a novel strategy for the treatment of liver fibrosis in patients.
Collapse
Affiliation(s)
- Hyun Young Kim
- Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Surgery, University of California San Diego, La Jolla, California, USA
| | - Sara Brin Rosenthal
- Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, California, USA
| | - Xiao Liu
- Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Surgery, University of California San Diego, La Jolla, California, USA
| | - Charlene Miciano
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA; Center for Epigenomics, University of California San Diego, La Jolla, California, USA
| | - Xiaomeng Hou
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA; Center for Epigenomics, University of California San Diego, La Jolla, California, USA
| | - Michael Miller
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA; Center for Epigenomics, University of California San Diego, La Jolla, California, USA
| | - Justin Buchanan
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA; Center for Epigenomics, University of California San Diego, La Jolla, California, USA
| | - Olivier B Poirion
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA; Center for Epigenomics, University of California San Diego, La Jolla, California, USA
| | - Daisy Chilin-Fuentes
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Cuijuan Han
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Mojgan Housseini
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Raquel Carvalho-Gontijo Weber
- Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Surgery, University of California San Diego, La Jolla, California, USA
| | - Sadatsugu Sakane
- Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Surgery, University of California San Diego, La Jolla, California, USA
| | - Wonseok Lee
- Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Surgery, University of California San Diego, La Jolla, California, USA
| | - Huayi Zhao
- Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Surgery, University of California San Diego, La Jolla, California, USA
| | - Karin Diggle
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Sebastian Preissl
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA; Center for Epigenomics, University of California San Diego, La Jolla, California, USA; Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA; Center for Epigenomics, University of California San Diego, La Jolla, California, USA
| | - Allen Wang
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA; Center for Epigenomics, University of California San Diego, La Jolla, California, USA.
| | - David A Brenner
- Department of Medicine, University of California San Diego, La Jolla, California, USA; Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
6
|
Shang J, Zhou C, He M, Huang XY, Qin CF, Wu A. Mutation S139N on Zika virus prM protein shifts immune response from Asian to contemporary strain. Brain Behav Immun 2025; 126:247-259. [PMID: 39986659 DOI: 10.1016/j.bbi.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 01/26/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025] Open
Abstract
Zika virus (ZIKV) has been associated with neurological diseases like microcephaly and Guillain-Barré syndrome. The S139N single mutation on the prM protein of the FSS13025 Asian strain increases the mortality rate in mice. Therefore, it is a valuable tool for studying the impact of immune responses on neural damage. Here, we used single-cell sequencing technology to systematically assess the immune response induced by three ZIKV strains: Asian ancestral strain FSS13025/2010, FSS13025 strain with S139N mutation (FSS13025-S139N), and contemporary strain GZ01/2016. By infecting 1-day-old mice, we observed that the immune spectrum elicited by FSS13025-S139N mutant resembled that induced by the contemporary strain. The FSS13025-S139N strain induces the proliferation of inflammatory microglial cells earlier than the FSS13025 strain, similar to GZ01. A specific cell cluster, Microglia_Ccr7, was induced by the S139N mutant strain and GZ01 strain, which suppresses T cell activation through the PDCD1LG2-PDCD1 signaling pathway. Furthermore, the proliferation of CD8+ T cells was weakened and prolonged in S139N strain-infected samples. Finally, we found that the S139N mutant strain causes more apoptosis of neurons compared to the FSS13025 strain. These results indicate that the S139N mutation plays an important role in the immune response pattern of ZIKV and prolongs the duration of neuroinflammation.
Collapse
Affiliation(s)
- Jingzhe Shang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123 Jiangsu, China; Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Chao Zhou
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China
| | - Mengjiao He
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China
| | - Xing-Yao Huang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China.
| | - Aiping Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123 Jiangsu, China; Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
| |
Collapse
|
7
|
Wang G, Li G, Song A, Zhao Y, Yu J, Wang Y, Dai W, Salas M, Qin H, Medrano L, Dow J, Li A, Armstrong B, Fueger PT, Yu H, Zhu Y, Shao M, Wu X, Jiang L, Campisi J, Yang X, Wang QA. Distinct adipose progenitor cells emerging with age drive active adipogenesis. Science 2025; 388:eadj0430. [PMID: 40273250 DOI: 10.1126/science.adj0430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 05/30/2024] [Accepted: 02/05/2025] [Indexed: 04/26/2025]
Abstract
Starting at middle age, adults often suffer from visceral adiposity and associated adverse metabolic disorders. Lineage tracing in mice revealed that adipose progenitor cells (APCs) in visceral fat undergo extensive adipogenesis during middle age. Thus, despite the low turnover rate of adipocytes in young adults, adipogenesis is unlocked during middle age. Transplantations quantitatively showed that APCs in middle-aged mice exhibited high adipogenic capacity cell-autonomously. Single-cell RNA sequencing identified a distinct APC population, the committed preadipocyte, age-enriched (CP-A), emerging at this age. CP-As demonstrated elevated proliferation and adipogenesis activity. Pharmacological and genetic manipulations indicated that leukemia inhibitory factor receptor signaling was indispensable for CP-A adipogenesis and visceral fat expansion. These findings uncover a fundamental mechanism of age-dependent adipose remodeling, offering critical insights into age-related metabolic diseases.
Collapse
Affiliation(s)
- Guan Wang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Gaoyan Li
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anying Song
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Yutian Zhao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jiayu Yu
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Yifan Wang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Wenting Dai
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Martha Salas
- Light Microscopy Core, City of Hope Medical Center, Duarte, CA, USA
| | - Hanjun Qin
- The Integrative Genomics Core, City of Hope Medical Center, Duarte, CA, USA
| | - Leonard Medrano
- Division of Developmental and Translational Diabetes and Endocrinology Research, City of Hope Medical Center, Duarte, CA, USA
| | - Joan Dow
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Metabolic Phenotyping Core, City of Hope Medical Center, Duarte, CA, USA
| | - Aimin Li
- Pathology Core of Shared Resources, City of Hope Medical Center, Duarte, CA, USA
| | - Brian Armstrong
- Light Microscopy Core, City of Hope Medical Center, Duarte, CA, USA
| | - Patrick T Fueger
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Metabolic Phenotyping Core, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Hua Yu
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | - Yi Zhu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Mengle Shao
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Xiwei Wu
- The Integrative Genomics Core, City of Hope Medical Center, Duarte, CA, USA
| | - Lei Jiang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| | | | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Qiong A Wang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Medical Center, Duarte, CA, USA
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, USA
| |
Collapse
|
8
|
Van Nerum K, Wenzel A, Argemi-Muntadas L, Kafkia E, Drews A, Brun IS, Lavro V, Roelofsen A, Stamidis N, Arnal SB, Zhao C, di Sanzo S, Völker-Albert M, Petropoulos S, Moritz T, Żylicz JJ. α-Ketoglutarate promotes trophectoderm induction and maturation from naive human embryonic stem cells. Nat Cell Biol 2025:10.1038/s41556-025-01658-1. [PMID: 40269259 DOI: 10.1038/s41556-025-01658-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/21/2025] [Indexed: 04/25/2025]
Abstract
Development and lineage choice are driven by interconnected transcriptional, epigenetic and metabolic changes. Specific metabolites, such as α-ketoglutarate (αKG), function as signalling molecules affecting the activity of chromatin-modifying enzymes. However, how metabolism coordinates cell-state changes, especially in human pre-implantation development, remains unclear. Here we uncover that inducing naive human embryonic stem cells towards the trophectoderm lineage results in considerable metabolic rewiring, characterized by αKG accumulation. Elevated αKG levels potentiate the capacity of naive embryonic stem cells to specify towards the trophectoderm lineage. Moreover, increased αKG levels promote blastoid polarization and trophectoderm maturation. αKG supplementation does not affect global histone methylation levels; rather, it decreases acetyl-CoA availability, reduces histone acetyltransferase activity and weakens the pluripotency network. We propose that metabolism functions as a positive feedback loop aiding in trophectoderm fate induction and maturation, highlighting that global metabolic rewiring can promote specificity in cell fate decisions through intricate regulation of signalling and chromatin.
Collapse
Affiliation(s)
- Karlien Van Nerum
- Novo Nordisk Foundation Center for Stem Cell Medicine - reNEW, Department of Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Anne Wenzel
- Novo Nordisk Foundation Center for Stem Cell Medicine - reNEW, Department of Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Lidia Argemi-Muntadas
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Eleni Kafkia
- Novo Nordisk Foundation Center for Stem Cell Medicine - reNEW, Department of Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Antar Drews
- Novo Nordisk Foundation Center for Stem Cell Medicine - reNEW, Department of Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Ida Sophie Brun
- Novo Nordisk Foundation Center for Stem Cell Medicine - reNEW, Department of Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Viktoria Lavro
- Novo Nordisk Foundation Center for Stem Cell Medicine - reNEW, Department of Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Annina Roelofsen
- Novo Nordisk Foundation Center for Stem Cell Medicine - reNEW, Department of Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Nikolaos Stamidis
- Novo Nordisk Foundation Center for Stem Cell Medicine - reNEW, Department of Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Sandra Bages Arnal
- Novo Nordisk Foundation Center for Stem Cell Medicine - reNEW, Department of Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Cheng Zhao
- Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Sophie Petropoulos
- Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Axe Immunopathologie, Montreal, Quebec, Canada
- Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas Moritz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jan Jakub Żylicz
- Novo Nordisk Foundation Center for Stem Cell Medicine - reNEW, Department of Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
9
|
Azami T, Theeuwes B, Nu Ton ML, Mansfield W, Harland L, Kinoshita M, Gottgens B, Nichols J. STAT3 signaling enhances tissue expansion during postimplantation mouse development. Cell Rep 2025; 44:115506. [PMID: 40188437 DOI: 10.1016/j.celrep.2025.115506] [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: 03/03/2024] [Revised: 01/10/2025] [Accepted: 03/12/2025] [Indexed: 04/08/2025] Open
Abstract
Signal transducer and activator of transcription (STAT)3 signaling has been studied extensively using mouse embryonic stem cells. Zygotic deletion of Stat3 enables embryo implantation, but thereafter, mutants resemble non-affected littermates from the previous day until around mid-gestation. This probably results from the loss of serine-phosphorylated STAT3, the predominant form in early postimplantation embryonic tissues associated with mitochondrial activity. Bulk RNA sequencing of isolated mouse epiblasts confirmed developmental delay transcriptionally. Single-cell RNA sequencing revealed the exclusion of derivatives of Stat3 null embryonic stem cells exclusively from the erythroid lineage of mid-gestation chimeras. We show that Stat3 null embryonic stem cells can differentiate into erythroid and hematopoietic lineages in vitro but become outcompeted when mixed with wild-type cells. Our results implicate a role for STAT3 in the temporal control of embryonic progression, particularly in tissues requiring rapid cell division, such as postimplantation epiblast and hematopoietic lineages. Interestingly, mutations in STAT3 are associated with short stature in humans.
Collapse
Affiliation(s)
- Takuya Azami
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK.
| | - Bart Theeuwes
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Mai-Linh Nu Ton
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - William Mansfield
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Luke Harland
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Masaki Kinoshita
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Berthold Gottgens
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Jennifer Nichols
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 3EG, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
10
|
Nano PR, Fazzari E, Azizad D, Martija A, Nguyen CV, Wang S, Giang V, Kan RL, Yoo J, Wick B, Haeussler M, Bhaduri A. Integrated analysis of molecular atlases unveils modules driving developmental cell subtype specification in the human cortex. Nat Neurosci 2025:10.1038/s41593-025-01933-2. [PMID: 40259073 DOI: 10.1038/s41593-025-01933-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/27/2025] [Indexed: 04/23/2025]
Abstract
Human brain development requires generating diverse cell types, a process explored by single-cell transcriptomics. Through parallel meta-analyses of the human cortex in development (seven datasets) and adulthood (16 datasets), we generated over 500 gene co-expression networks that can describe mechanisms of cortical development, centering on peak stages of neurogenesis. These meta-modules show dynamic cell subtype specificities throughout cortical development, with several developmental meta-modules displaying spatiotemporal expression patterns that allude to potential roles in cell fate specification. We validated the expression of these modules in primary human cortical tissues. These include meta-module 20, a module elevated in FEZF2+ deep layer neurons that includes TSHZ3, a transcription factor associated with neurodevelopmental disorders. Human cortical chimeroid experiments validated that both FEZF2 and TSHZ3 are required to drive module 20 activity and deep layer neuron specification but through distinct modalities. These studies demonstrate how meta-atlases can engender further mechanistic analyses of cortical fate specification.
Collapse
Affiliation(s)
- Patricia R Nano
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Elisa Fazzari
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daria Azizad
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Antoni Martija
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Claudia V Nguyen
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sean Wang
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vanna Giang
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ryan L Kan
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Juyoun Yoo
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Brittney Wick
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | | | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
11
|
May A, Röper K. Single-cell analysis of the early Drosophila salivary gland reveals that morphogenetic control involves both the induction and exclusion of gene expression programs. PLoS Biol 2025; 23:e3003133. [PMID: 40258079 DOI: 10.1371/journal.pbio.3003133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 03/25/2025] [Indexed: 04/23/2025] Open
Abstract
How tissue shape and therefore function is encoded by the genome remains in many cases unresolved. The tubes of the salivary glands in the Drosophila embryo start from simple epithelial placodes, specified through the homeotic factors Scr/Hth/Exd. Previous work indicated that early morphogenetic changes are prepatterned by transcriptional changes, but an exhaustive transcriptional blueprint driving physical changes was lacking. We performed single-cell-RNAseq-analysis of FACS-isolated early placodal cells, making up less than 0.4% of cells within the embryo. Differential expression analysis in comparison to epidermal cells analyzed in parallel generated a repertoire of genes highly upregulated within placodal cells prior to morphogenetic changes. Furthermore, clustering and pseudotime analysis of single-cell-sequencing data identified dynamic expression changes along the morphogenetic timeline. Our dataset provides a comprehensive resource for future studies of a simple but highly conserved morphogenetic process of tube morphogenesis. Unexpectedly, we identified a subset of genes that, although initially expressed in the very early placode, then became selectively excluded from the placode but not the surrounding epidermis, including hth, grainyhead and tollo/toll-8. We show that maintaining tollo expression severely compromised the tube morphogenesis. We propose tollo is switched off to not interfere with key Tolls/LRRs that are expressed and function in the tube morphogenesis.
Collapse
Affiliation(s)
- Annabel May
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Katja Röper
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
12
|
Daugelaite K, Lacour P, Winkler I, Koch ML, Schneider A, Schneider N, Coraggio F, Tolkachov A, Nguyen XP, Vilkaite A, Rehnitz J, Odom DT, Goncalves A. Granulosa cell transcription is similarly impacted by superovulation and aging and predicts early embryonic trajectories. Nat Commun 2025; 16:3658. [PMID: 40246835 PMCID: PMC12006393 DOI: 10.1038/s41467-025-58451-9] [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: 11/03/2023] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
In vitro fertilization efficiency is limited in part because a fraction of retrieved oocytes fails to fertilize. Accurately evaluating their quality could significantly improve in vitro fertilization efficiency, which would require better understanding how their maturation may be disrupted. Here, we quantitatively investigate the interplay between superovulation and aging in mouse oocytes and their paired granulosa cells using a newly adapted experimental methodology. We test the hypothesis that superovulation disrupts oocyte maturation, revealing the key intercellular communication pathways dysregulated at the transcriptional level by forced hormonal stimulation. We further demonstrate that granulosa cell transcriptional markers can prospectively predict an associated oocyte's early developmental potential. By using naturally ovulated old mice as a non-stimulated reference, we show that aging and superovulation dysregulate similar genes and interact with each other. By comparing mice and human transcriptional responses of granulosa cells, we find that age-related dysregulation of hormonal responses and cell cycle pathways are shared, though substantial divergence exists in other pathways.
Collapse
Affiliation(s)
- Klaudija Daugelaite
- Division of Regulatory Genomics and Cancer Evolution, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Ruprecht-Karl-University Heidelberg, Heidelberg, Germany
| | - Perrine Lacour
- Faculty of Biosciences, Ruprecht-Karl-University Heidelberg, Heidelberg, Germany
- Division of Molecular and Computational Prevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ivana Winkler
- Division of Molecular and Computational Prevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marie-Luise Koch
- Division of Regulatory Genomics and Cancer Evolution, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Schneider
- Division of Regulatory Genomics and Cancer Evolution, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nina Schneider
- Division of Molecular and Computational Prevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Francesca Coraggio
- Division of Molecular and Computational Prevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Tolkachov
- Division of Regulatory Genomics and Cancer Evolution, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cardiology, Angiology and Pneumology, Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Xuan Phuoc Nguyen
- Department of Gynecological Endocrinology and Fertility Disorders, University Women's Hospital Heidelberg, Heidelberg, Germany
| | - Adriana Vilkaite
- Department of Gynecological Endocrinology and Fertility Disorders, University Women's Hospital Heidelberg, Heidelberg, Germany
| | - Julia Rehnitz
- Department of Gynecological Endocrinology and Fertility Disorders, University Women's Hospital Heidelberg, Heidelberg, Germany
| | - Duncan T Odom
- Division of Regulatory Genomics and Cancer Evolution, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Angela Goncalves
- Division of Molecular and Computational Prevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
13
|
Wu Z, Wang Y, Chen WW, Sun H, Chen X, Li X, Wang Z, Liang W, Wang SY, Luan X, Li Y, Huang S, Liang Y, Zhang J, Chen ZF, Wang G, Gao Y, Liu Y, Wang J, Liu Z, Shi P, Liu C, Lv L, Hou A, Wu C, Yao C, Hong Z, Dai J, Lu Z, Pan F, Chen X, Kettenmann H, Amit I, Speakman JR, Chen Y, Ginhoux F, Cui R, Huang T, Li H. Peripheral nervous system microglia-like cells regulate neuronal soma size throughout evolution. Cell 2025; 188:2159-2174.e15. [PMID: 40199320 DOI: 10.1016/j.cell.2025.02.007] [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: 03/20/2024] [Revised: 11/18/2024] [Accepted: 02/12/2025] [Indexed: 04/10/2025]
Abstract
Microglia, essential in the central nervous system (CNS), were historically considered absent from the peripheral nervous system (PNS). Here, we show a PNS-resident macrophage population that shares transcriptomic and epigenetic profiles as well as an ontogenetic trajectory with CNS microglia. This population (termed PNS microglia-like cells) enwraps the neuronal soma inside the satellite glial cell envelope, preferentially associates with larger neurons during PNS development, and is required for neuronal functions by regulating soma enlargement and axon growth. A phylogenetic survey of 24 vertebrates revealed an early origin of PNS microglia-like cells, whose presence is correlated with neuronal soma size (and body size) rather than evolutionary distance. Consistent with their requirement for soma enlargement, PNS microglia-like cells are maintained in vertebrates with large peripheral neuronal soma but absent when neurons evolve to have smaller soma. Our study thus reveals a PNS counterpart of CNS microglia that regulates neuronal soma size during both evolution and ontogeny.
Collapse
Affiliation(s)
- Zhisheng Wu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Department of Immunology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; School of Chemistry and Chemical Engineering, Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yiheng Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wei-Wei Chen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hua Sun
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; School of Life Sciences, Henan University, Henan, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Xiaobo Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zeshuai Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weizheng Liang
- Hebei Provincial Key Laboratory of Systems Biology and Gene Regulation, Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Shuang-Yin Wang
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Xuemei Luan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yijiang Li
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Shangjin Huang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuteng Liang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jiaqi Zhang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhou-Feng Chen
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, and Shenzhen Medical Academy of Research and Translation, Shenzhen, China
| | - Guanlin Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China; Shanghai Qi Zhi Institute, Shanghai, China
| | - Yun Gao
- State Key Laboratory of Genetic Resources and Evolution, and Southwest Research Centre of Porcine Molecular Breeding and Translational Medicine in China, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yanan Liu
- State Key Laboratory of Genetic Resources and Evolution, and Southwest Research Centre of Porcine Molecular Breeding and Translational Medicine in China, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jun Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Liu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Peng Shi
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Cirong Liu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Longbao Lv
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Anli Hou
- Shenzhen Guangming District People's Hospital, Shenzhen, China
| | - Chenglin Wu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chen Yao
- The First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, Shenzhen, China
| | - Zexuan Hong
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ji Dai
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhonghua Lu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fan Pan
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | | | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Yun Chen
- Department of Immunology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; School of Chemistry and Chemical Engineering, Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| | - Florent Ginhoux
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Rongfeng Cui
- School of Ecology & State Key Laboratory of Biocontrol, Sun Yat-sen University, Shenzhen, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, China
| | - Tianwen Huang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hanjie Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen University of Advanced Technology, Shenzhen, China.
| |
Collapse
|
14
|
Wang J, Zhou Y, Zhang M, Li X, Liu T, Liu Y, Xie H, Wang K, Li P, Xu Z, Duan B. Resolving floral development dynamics using genome and single-cell temporal transcriptome of Dendrobium devonianum. PLANT BIOTECHNOLOGY JOURNAL 2025. [PMID: 40238860 DOI: 10.1111/pbi.70094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
Dendrobium devonianum, a species of the Orchidaceae family, is notable for its unique floral characteristics, which include two yellow spots and purple tips on its labellum, as well as fringed edges. However, the molecular mechanisms underlying flower pattern formation in D. devonianum remain poorly understood, hindering advancements in its breeding process. Here, a chromosome-scale genome of D. devonianum was presented for the first time, revealing two significant polyploidization events. Additionally, a high-resolution single-cell transcriptomic atlas was constructed, capturing 11 distinct cell clusters. Expression patterns of MADS-box genes were identified through temporal and spatial bulk RNA-Seq, revealing alignment with the ABCDE model of flower formation. Meanwhile, mass spectrometry imaging and scRNA analyses showed that the yellow spots were primarily associated with carotenoid biosynthesis gene expression, while the purple colour is predominantly linked to anthocyanin biosynthesis gene expression. These genes were mainly expressed in the epidermis and vascular cells. Developmental trajectory analyses of epidermal cells further uncovered a gene regulatory network and several transcription factors likely responsible for fringes formation along the labellum margin. This study provides valuable insights into the molecular mechanisms driving floral colour differentiation and structural traits in D. devonianum, contributing to a deeper understanding of orchid evolution, diversification and breeding.
Collapse
Affiliation(s)
- Jing Wang
- College of Pharmaceutical Science, Dali University, Dali, China
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Ying Zhou
- College of Pharmaceutical Science, Dali University, Dali, China
- Institute of Caulis Dendrobii Longling County, Baoshan, China
| | - Manchang Zhang
- Institute of Caulis Dendrobii Longling County, Baoshan, China
- International Joint Laboratory for the Development and Utilization of Traditional Chinese Medicine Resources in Yunnan Province, Baoshan, Dali, China
- Baoshan Food and Drug Inspection and Testing Center, Baoshan, China
| | - Xinping Li
- College of Pharmaceutical Science, Dali University, Dali, China
- College of Life Science, Northeast Forestry University, Harbin, China
- International Joint Laboratory for the Development and Utilization of Traditional Chinese Medicine Resources in Yunnan Province, Baoshan, Dali, China
| | - Tingxia Liu
- College of Pharmaceutical Science, Dali University, Dali, China
- International Joint Laboratory for the Development and Utilization of Traditional Chinese Medicine Resources in Yunnan Province, Baoshan, Dali, China
| | - Yinglin Liu
- College of Pharmaceutical Science, Dali University, Dali, China
- International Joint Laboratory for the Development and Utilization of Traditional Chinese Medicine Resources in Yunnan Province, Baoshan, Dali, China
| | - He Xie
- Tobacco Breeding and Biotechnology Research Center, Yunnan Academy of Tobacco Agricultural Sciences, Kunming, Yunnan, China
| | - Kaiying Wang
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Peng Li
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Zhichao Xu
- College of Life Science, Northeast Forestry University, Harbin, China
- International Joint Laboratory for the Development and Utilization of Traditional Chinese Medicine Resources in Yunnan Province, Baoshan, Dali, China
| | - Baozhong Duan
- College of Pharmaceutical Science, Dali University, Dali, China
- College of Life Science, Northeast Forestry University, Harbin, China
- International Joint Laboratory for the Development and Utilization of Traditional Chinese Medicine Resources in Yunnan Province, Baoshan, Dali, China
| |
Collapse
|
15
|
Cao K, Wei S, Ma T, Yang X, Wang Y, He X, Lu M, Bai Y, Qi C, Zhang L, Li L, Meng H, Ma J, Zhu J. Integrating bulk, single-cell, and spatial transcriptomics to identify and functionally validate novel targets to enhance immunotherapy in NSCLC. NPJ Precis Oncol 2025; 9:112. [PMID: 40240582 PMCID: PMC12003664 DOI: 10.1038/s41698-025-00893-x] [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: 10/20/2024] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Programmed cell deaths (PCDs) are crucial for tumor progression. By analyzing 18 PCDs, we generated a robust multigene signature, Combined Cell Death Index (CCDI), comprising necroptosis and autophagy genes for non-small cell lung cancer (NSCLC). The CCDI accurately stratified patients by survival prognosis and predicted immunotherapy responses. We validated CCDI and prioritized CCDI genes using five single-cell RNA sequencing and two spatial transcriptomics datasets. CCDI positively correlates with tumor malignancy, invasiveness, and immunotherapy resistance. Four necroptosis genes (PTGES3, MYO6, CCT6A, and CTSH) may affect cancer cell evolution. In vitro, CTSH overexpression or PTGES3 knockdown inhibited NSCLC cell proliferation and migration while inducing necroptosis with necrosome formation. Moreover, we observed diminished CTSH, heightened PTGES3, and low necroptosis activity in 12 pairs of NSCLC tumors and normal tissues. CTSH overexpression or PTGES3 knockdown induced necroptosis and improved anti-PD1 therapy efficiency in syngeneic cancer mouse models. These findings indicate necroptosis genes as potential therapeutic targets in cancer treatments.
Collapse
Affiliation(s)
- Kui Cao
- Department of Thoracic Surgery, Harbin Medical University Cancer hospital, Harbin, Heilongjiang, China
| | - Shenshui Wei
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Tianjiao Ma
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinxin Yang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yuning Wang
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Xiangrong He
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Mengdi Lu
- Department of Thoracic Surgery, Harbin Medical University Cancer hospital, Harbin, Heilongjiang, China
| | - Yuwen Bai
- Department of Thoracic Surgery, Harbin Medical University Cancer hospital, Harbin, Heilongjiang, China
| | - Cuicui Qi
- Department of Thoracic Surgery, Harbin Medical University Cancer hospital, Harbin, Heilongjiang, China
| | - Luquan Zhang
- Department of Thoracic Surgery, Harbin Medical University Cancer hospital, Harbin, Heilongjiang, China
| | - Lijuan Li
- Department of Thoracic Surgery, Harbin Medical University Cancer hospital, Harbin, Heilongjiang, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| | - Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer hospital, Harbin, Heilongjiang, China.
| | - Jinhong Zhu
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
- Biobank, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| |
Collapse
|
16
|
Xie Y, Jing Z, Pan H, Xu X, Fang Q. Redefining the high variable genes by optimized LOESS regression with positive ratio. BMC Bioinformatics 2025; 26:104. [PMID: 40234751 PMCID: PMC12001687 DOI: 10.1186/s12859-025-06112-5] [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: 11/18/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Single-cell RNA sequencing allows for the exploration of transcriptomic features at the individual cell level, but the high dimensionality and sparsity of the data pose substantial challenges for downstream analysis. Feature selection, therefore, is a critical step to reduce dimensionality and enhance interpretability. RESULTS We developed a robust feature selection algorithm that leverages optimized locally estimated scatterplot smoothing regression (LOESS) to precisely capture the relationship between gene average expression level and positive ratio while minimizing overfitting. Our evaluations showed that our algorithm consistently outperforms eight leading feature selection methods across three benchmark criteria and helps improve downstream analysis, thus offering a significant improvement in gene subset selection. CONCLUSIONS By preserving key biological information through feature selection, GLP provides informative features to enhance the accuracy and effectiveness of downstream analyses.
Collapse
Affiliation(s)
- Yue Xie
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Zehua Jing
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | | | - Xun Xu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- BGI Research, Shenzhen, 518083, China.
| | - Qi Fang
- BGI Research, Shenzhen, 518083, China.
| |
Collapse
|
17
|
Blaszczyk GJ, Piscopo VEC, Goldsmith TM, Chapleau A, Sirois J, Bernard G, Antel JP, Durcan TM. Single cell RNAseq to identify subpopulations of glial progenitors in iPSC-derived oligodendroglial lineage cultures. NPJ Syst Biol Appl 2025; 11:35. [PMID: 40234453 PMCID: PMC12000351 DOI: 10.1038/s41540-025-00515-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: 12/18/2024] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
Cellular heterogeneity is a common issue in differentiation protocols of oligodendrocytes (OLs) from human induced pluripotent stem cells. Our previous work described a novel method to generate OLs and highlighted the presence of glial progenitors. Here, we unravel the glial heterogeneity and characterize the response of isolated subpopulations to differentiation. This study provides a novel tool for studying the dynamics of glial development in vitro and on a transcriptomic level.
Collapse
Affiliation(s)
- Gabriela J Blaszczyk
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada, QC
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada, QC
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada, QC
| | - Valerio E C Piscopo
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada, QC
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada, QC
| | - Taylor M Goldsmith
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada, QC
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada, QC
| | - Alexandra Chapleau
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada, QC
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada, QC
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, Canada, QC
| | - Julien Sirois
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada, QC
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada, QC
| | - Geneviève Bernard
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada, QC
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, Canada, QC
- Department of Pediatrics and Human Genetics, McGill University, Montreal, Canada, QC
- Division of Medical Genetics, Department of Specialized Medicine, McGill University Health Centre, Montreal, Canada, QC
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada, QC
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada, QC
| | - Thomas M Durcan
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada, QC.
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada, QC.
| |
Collapse
|
18
|
Li H, Zandberg DP, Kulkarni A, Chiosea SI, Santos PM, Isett BR, Joy M, Sica GL, Contrera KJ, Tatsuoka CM, Brand M, Duvvuri U, Kim S, Kubik M, Sridharan S, Tu F, Chen J, Bruno TC, Vignali DAA, Cillo AR, Bao R, Wang JH, Vujanovic L, Ferris RL. Distinct CD8 + T cell dynamics associate with response to neoadjuvant cancer immunotherapies. Cancer Cell 2025; 43:757-775.e8. [PMID: 40086437 DOI: 10.1016/j.ccell.2025.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/30/2024] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
We leverage a clinical trial (NCT04080804) that compared neoadjuvant anti-PD-1, anti-PD-1+CTLA-4, and anti-PD-1+LAG-3 therapies in head and neck squamous cell carcinoma patients. Combination therapies promote higher pathologic response rates versus monotherapy, and major pathologic response is associated with better survival. To address whether successful immune checkpoint inhibitor (ICI) regimens act through similar or distinct pathways, we robustly and longitudinally characterize transcriptional and proteomic dynamics of CD8+ tumor-infiltrating lymphocytes (TILs) in a clonal manner. Anti-PD-1+LAG-3 reprograms CD8+ TIL with type-I interferon response and exhaustion gene programs into effector memory and resident memory (TEM/TRM). In contrast, anti-PD-1+CTLA-4 activates and expands pre-existing TEM/TRM CD8+ TIL, but does not rejuvenate exhausted phenotypes into T effector cells. Anti-PD-1+LAG-3, but not anti-PD-1+CTLA-4, induces widespread TCR sharing among the different transcriptional states, as well as increased TCR diversity in responding patients. Our data suggest doublet regimen-specific transcriptional and clonal dynamics of tumor-reactive CD8+ T cells.
Collapse
Affiliation(s)
- Housaiyin Li
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Molecular Genetics and Development Biology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dan P Zandberg
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Aditi Kulkarni
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Simion I Chiosea
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patricia M Santos
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian R Isett
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marion Joy
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Gabriel L Sica
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin J Contrera
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Curtis M Tatsuoka
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Biostatistics Facility, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Matthias Brand
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Umamaheswar Duvvuri
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, NYU Grossman School of Medicine, New York, NY, USA
| | - Seungwon Kim
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark Kubik
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shaum Sridharan
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fei Tu
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jie Chen
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Molecular Genetics and Development Biology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tullia C Bruno
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Dario A A Vignali
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Anthony R Cillo
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Riyue Bao
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jing Hong Wang
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Lazar Vujanovic
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Robert L Ferris
- UNC Lineberger Comprehensive Cancer Center, UNC Health Care System, Chapel Hill, NC, USA.
| |
Collapse
|
19
|
Yates J, Van Allen EM. New horizons at the interface of artificial intelligence and translational cancer research. Cancer Cell 2025; 43:708-727. [PMID: 40233719 PMCID: PMC12007700 DOI: 10.1016/j.ccell.2025.03.018] [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: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 04/17/2025]
Abstract
Artificial intelligence (AI) is increasingly being utilized in cancer research as a computational strategy for analyzing multiomics datasets. Advances in single-cell and spatial profiling technologies have contributed significantly to our understanding of tumor biology, and AI methodologies are now being applied to accelerate translational efforts, including target discovery, biomarker identification, patient stratification, and therapeutic response prediction. Despite these advancements, the integration of AI into clinical workflows remains limited, presenting both challenges and opportunities. This review discusses AI applications in multiomics analysis and translational oncology, emphasizing their role in advancing biological discoveries and informing clinical decision-making. Key areas of focus include cellular heterogeneity, tumor microenvironment interactions, and AI-aided diagnostics. Challenges such as reproducibility, interpretability of AI models, and clinical integration are explored, with attention to strategies for addressing these hurdles. Together, these developments underscore the potential of AI and multiomics to enhance precision oncology and contribute to advancements in cancer care.
Collapse
Affiliation(s)
- Josephine Yates
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland; ETH AI Center, ETH Zurich, Zurich, Switzerland; Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Medical Sciences, Harvard University, Boston, MA, USA; Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
20
|
Hao T, Pei Z, Hu S, Zhao Z, He W, Wang J, Jiang L, Ariben J, Wu L, Yang X, Wang L, Wu Y, Chen X, Li Q, Yang H, Li S, Wang X, Sun M, Zhang B. Identification of osteoarthritis-associated chondrocyte subpopulations and key gene-regulating drugs based on multi-omics analysis. Sci Rep 2025; 15:12448. [PMID: 40216809 PMCID: PMC11992032 DOI: 10.1038/s41598-025-90694-w] [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: 10/08/2024] [Accepted: 02/14/2025] [Indexed: 04/14/2025] Open
Abstract
The mechanism by which chondrocytes respond to mechanical stress in joints significantly affects the balance and function of cartilage. This study aims to characterize osteoarthritis-associated chondrocyte subpopulations and key gene targets for regulatory drugs. To begin, single-cell and transcriptome datasets were obtained from the Gene Expression Omnibus (GEO) database. Cell communication and pseudo-temporal analysis, as well as High-dimensional Weighted Gene Co-expression Network Analysis (hdWGCNA), were conducted on the single-cell data to identify key chondrocyte subtypes and module genes. Subsequently, Consensus Cluster Plus analysis was utilized to identify distinct disease subgroups within the osteoarthritis (OA) training dataset based on the key module genes. Furthermore, differential gene expression analysis and GO/KEGG pathway enrichment analysis were performed on the identified subgroups. To screen for hub genes associated with OA, a combination of 10 machine learning algorithms and 113 algorithm compositions was integrated. Additionally, the immune and pathway scores of the training dataset samples were evaluated using the ESTIMATE, MCP-counter, and ssGSEA algorithms to establish the relationship between the hub genes and immune and pathways. Following this, a network depicting the interaction between the hub genes and transcription factors was constructed based on the Network Analyst database. Moreover, the hub genes were subjected to drug prediction and molecular docking using the RNAactDrug database and AutoDockTools. Finally, real-time fluorescence quantitative PCR (RT-qPCR) was employed to detect the expression of hub genes in the plasma samples collected from osteoarthritis patients and healthy adults. In the OA sample, there is a significant increase in the proportion of prehypertrophic chondrocytes (preHTC), particularly in subgroups 6, 7, and 9. We defined these subgroups as OA_PreHTC subgroups. The OA_PreHTC subgroup exhibits a higher communication intensity with proliferative-related pathways such as ANGPTL and TGF-β. Furthermore, two OA disease subgroups were identified in the training set samples. This led to the identification of 411 differentially expressed genes (DEGs) related to osteoarthritis, 2485 DEGs among subgroups, as well as 238 intersecting genes and 5 hub genes (MMP13, FAM26F, CHI3L1, TAC1, and CKS2). RT-qPCR results indicate significant differences in the expression levels of five hub genes and their related TFs in the clinical blood samples of OA patients compared to the healthy control group (NC). Moreover, these five hub genes are positively associated with inflammatory pathways such as TNF-α, JAK-STAT3, and inflammatory response, while being negatively associated with proliferation pathways like WNT and KRAS. Additionally, the five hub genes are positively associated with neutrophils, activated CD4 T cell, gamma delta T cell, and regulatory T cell, while being negatively associated with CD56dim natural killer cell and Type 17T helper cell. Molecular docking results reveal that CAY10603, Tenulin, T0901317, and Nonactin exhibit high binding activity to CHI3L1, suggesting their potential as therapeutic drugs for OA. The OA_PreHTC subgroups plays a crucial role in the occurrence and development of osteoarthritis (OA). Five hub genes may exert their effects on OA through interactions with PreHTC cells, other chondrocytes, and immune cells, playing a role in inhibiting cell proliferation and stimulating inflammation, thus having high diagnostic value for OA. Additionally, CAY10603, Tenulin, T0901317, and Nonactin have potential therapeutic effects for OA patients.
Collapse
Affiliation(s)
- Ting Hao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Zhiwei Pei
- Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, People's Republic of China
| | - Sile Hu
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Zhenqun Zhao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Wanxiong He
- Sanya People's Hospital, No. 558 Jiefang Road, Sanya City, Hainan Province, People's Republic of China
| | - Jing Wang
- Baotou Medical College Bayannur Clinical Medical College, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Liuchang Jiang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Jirigala Ariben
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Lina Wu
- Aier Eye Hospital, Tianjin University, No. 102 Fukang Road, Tianjin, 300000, People's Republic of China
| | - Xiaolong Yang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Leipeng Wang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Yonggang Wu
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Xiaofeng Chen
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Qiang Li
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Haobo Yang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Siqin Li
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Xing Wang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China.
| | - Mingqi Sun
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
| | - Baoxin Zhang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
- Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, People's Republic of China.
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
| |
Collapse
|
21
|
Pentimalli TM, Schallenberg S, León-Periñán D, Legnini I, Theurillat I, Thomas G, Boltengagen A, Fritzsche S, Nimo J, Ruff L, Dernbach G, Jurmeister P, Murphy S, Gregory MT, Liang Y, Cordenonsi M, Piccolo S, Coscia F, Woehler A, Karaiskos N, Klauschen F, Rajewsky N. Combining spatial transcriptomics and ECM imaging in 3D for mapping cellular interactions in the tumor microenvironment. Cell Syst 2025:101261. [PMID: 40220761 DOI: 10.1016/j.cels.2025.101261] [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: 07/12/2024] [Revised: 12/13/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
Tumors are complex ecosystems composed of malignant and non-malignant cells embedded in a dynamic extracellular matrix (ECM). In the tumor microenvironment, molecular phenotypes are controlled by cell-cell and ECM interactions in 3D cellular neighborhoods (CNs). While their inhibition can impede tumor progression, routine molecular tumor profiling fails to capture cellular interactions. Single-cell spatial transcriptomics (ST) maps receptor-ligand interactions but usually remains limited to 2D tissue sections and lacks ECM readouts. Here, we integrate 3D ST with ECM imaging in serial sections from one clinical lung carcinoma to systematically quantify molecular states, cell-cell interactions, and ECM remodeling in CN. Our integrative analysis pinpointed known immune escape and tumor invasion mechanisms, revealing several druggable drivers of tumor progression in the patient under study. This proof-of-principle study highlights the potential of in-depth CN profiling in routine clinical samples to inform microenvironment-directed therapies. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Tancredi Massimo Pentimalli
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Daniel León-Periñán
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ivano Legnini
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Human Technopole, Milan, Italy
| | - Ilan Theurillat
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Gwendolin Thomas
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anastasiya Boltengagen
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sonja Fritzsche
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | - Jose Nimo
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | | | - Gabriel Dernbach
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany; Aignostics GmbH, Berlin, Germany; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | | | | | | | - Yan Liang
- NanoString® Technologies, Inc, Seattle, WA, USA
| | | | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Fabian Coscia
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Andrew Woehler
- Systems Biology Imaging Platform, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - Nikos Karaiskos
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Frederick Klauschen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany; Institute of Pathology, Ludwig Maximilians Universität, Munich, Germany
| | - Nikolaus Rajewsky
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany; NeuroCure Cluster of Excellence, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany; National Center for Tumor Diseases (NCT), Site Berlin, Berlin, Germany.
| |
Collapse
|
22
|
Batho-Samblas C, Smith J, Keavey L, Clancy N, McTeir L, Davey MG. Characterisation of the avascular mesenchyme during digit outgrowth. Dev Biol 2025; 523:99-110. [PMID: 40210155 DOI: 10.1016/j.ydbio.2025.04.005] [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: 10/30/2024] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
The avascular mesenchyme at the tip of the developing digit contributes to digit outgrowth and patterning, however, it has been poorly characterised. Using newly developed fate mapping approaches, tissue manipulation and single-cell mRNA sequencing data, we explore the transcriptional nature and developmental potential of this tissue. We find that the avascular mesenchyme is essential to normal segmental patterning of the digit and has a distinct transcriptional identity. In addition, we uncover an unexpected relationship between the unspecified tissue of the avascular mesenchyme and the committed phalanx forming region, which controls patterning, but not outgrowth of the digit. This multifaceted approach provides insights into the mechanics and genetic pathways that regulate digit outgrowth and patterning.
Collapse
Affiliation(s)
- Cameron Batho-Samblas
- Functional Genetics, The Roslin Institute & R(D)SVS, University of Edinburgh, EH25 9RG, UK
| | - Jonathan Smith
- Functional Genetics, The Roslin Institute & R(D)SVS, University of Edinburgh, EH25 9RG, UK; MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Lois Keavey
- Functional Genetics, The Roslin Institute & R(D)SVS, University of Edinburgh, EH25 9RG, UK; UK Dementia Research Institute, University of Edinburgh, EH16 4SB, UK
| | - Noah Clancy
- Functional Genetics, The Roslin Institute & R(D)SVS, University of Edinburgh, EH25 9RG, UK
| | - Lynn McTeir
- Functional Genetics, The Roslin Institute & R(D)SVS, University of Edinburgh, EH25 9RG, UK
| | - Megan G Davey
- Functional Genetics, The Roslin Institute & R(D)SVS, University of Edinburgh, EH25 9RG, UK; RICE- Roslin Institute Chicken Embryology, UK.
| |
Collapse
|
23
|
Benmebarek MR, Oguz C, Seifert M, Ruf B, Myojin Y, Bauer KC, Huang P, Ma C, Villamor-Payà M, Rodriguez-Matos F, Soliman M, Trehan R, Monge C, Xie C, Kleiner DE, Wood BJ, Levy EB, Budhu A, Kedei N, Mayer CT, Wang XW, Lack J, Telford W, Korangy F, Greten TF. Anti-vascular endothelial growth factor treatment potentiates immune checkpoint blockade through a BAFF- and IL-12-dependent reprogramming of the TME. Immunity 2025; 58:926-945.e10. [PMID: 40088889 PMCID: PMC11981852 DOI: 10.1016/j.immuni.2025.02.017] [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: 05/08/2024] [Revised: 10/31/2024] [Accepted: 02/12/2025] [Indexed: 03/17/2025]
Abstract
Anti-vascular endothelial growth factor (VEGF) treatment has shown clinical activity together with immune checkpoint blockade (ICB), but the exact mechanism is not known. We show that VEGF blockade in combination with anti-cytotoxic T-lymphocyte associated protein 4 (CTLA4) + anti-programmed death-ligand 1 (PD-L1) in cholangiocarcinoma (CCA) potentiated a multimodal mechanism dependent on B cell activating factor (BAFF), leading to a proinflammatory B cell response. It led to a BAFF- and interleukin (IL)-12-dependent expansion and rewiring of T regulatory cells (Tregs) toward an anti-tumor T helper-1 (Th-1)-like fragile state. We translated this approach to the clinic and observed immunological changes characterized by Treg cell expansion and rewiring toward fragile and unstable states. We explored the effect of VEGF receptor 2 (VEGFR2) signaling on Treg cell transcriptional programming and established a mouse model ablating VEGFR2 expression on Treg cells. This study reveals the immunological interplay resulting from targeting VEGF together with CTLA-4 and PD-L1 blockade.
Collapse
Affiliation(s)
- Mohamed-Reda Benmebarek
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Matthias Seifert
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Ruf
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yuta Myojin
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kylynda C Bauer
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Patrick Huang
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chi Ma
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marina Villamor-Payà
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Francisco Rodriguez-Matos
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marlaine Soliman
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rajiv Trehan
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cecilia Monge
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Changqing Xie
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - David E Kleiner
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bradford J Wood
- Center for Interventional Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elliot B Levy
- Center for Interventional Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anuradha Budhu
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Noemi Kedei
- CCR Collaborative Bioinformatics Resource, Office of Science and Technology Resources, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Christian T Mayer
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xin Wei Wang
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Justin Lack
- Integrated Data Sciences Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - William Telford
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Firouzeh Korangy
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
24
|
Molteni R, Fiumara M, Campochiaro C, Alfieri R, Pacini G, Licari E, Tomelleri A, Diral E, Varesi A, Weber A, Quaranta P, Albano L, Gaddoni C, Basso-Ricci L, Stefanoni D, Alessandrini L, Degl'Innocenti S, Sanvito F, Bergonzi GM, Annoni A, Panigada M, Cantoni E, Canarutto D, Xie SZ, D'Alessandro A, Di Micco R, Aiuti A, Ciceri F, De Luca G, Dagna L, Matucci-Cerinic M, Merelli I, Cenci S, Scala S, Cavalli G, Naldini L, Ferrari S. Mechanisms of hematopoietic clonal dominance in VEXAS syndrome. Nat Med 2025:10.1038/s41591-025-03623-9. [PMID: 40195449 DOI: 10.1038/s41591-025-03623-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/28/2025] [Indexed: 04/09/2025]
Abstract
Clonal dominance characterizes hematopoiesis during aging and increases susceptibility to blood cancers and common nonmalignant disorders. VEXAS syndrome is a recently discovered, adult-onset, autoinflammatory disease burdened by a high mortality rate and caused by dominant hematopoietic clones bearing somatic mutations in the UBA1 gene. However, pathogenic mechanisms driving clonal dominance are unknown. Moreover, the lack of disease models hampers the development of disease-modifying therapies. In the present study, we performed immunophenotype characterization of hematopoiesis and single-cell transcriptomics in a cohort of nine male patients with VEXAS syndrome, revealing pervasive inflammation across all lineages. Hematopoietic stem and progenitor cells (HSPCs) in patients are skewed toward myelopoiesis and acquire senescence-like programs. Humanized models of VEXAS syndrome, generated by inserting the causative mutation in healthy HSPCs through base editing, recapitulated proteostatic defects, cytological alterations and senescence signatures of patients' cells, as well as hematological and inflammatory disease hallmarks. Competitive transplantations of human UBA1-mutant and wild-type HSPCs showed that, although mutant cells are more resilient to the inflammatory milieu, probably through the acquisition of the senescence-like state, wild-type ones are progressively exhausted and overwhelmed by VEXAS clones, overall impairing functional hematopoiesis and leading to bone marrow failure. Our study unveils the mechanism of clonal dominance and provides models for preclinical studies and preliminary insights that could inform therapeutic strategies.
Collapse
Affiliation(s)
- Raffaella Molteni
- Vita-Salute San Raffaele University, Milan, Italy.
- Inflammation Fibrosis and Ageing Initiative (INFLAGE), Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Martina Fiumara
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Corrado Campochiaro
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Alfieri
- National Research Council, Institute for Biomedical Technologies, Segrate, Italy
| | - Guido Pacini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eugenia Licari
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Tomelleri
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Diral
- Unit of Hematology and Stem Cell Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angelica Varesi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Alessandra Weber
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pamela Quaranta
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luisa Albano
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Gaddoni
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Basso-Ricci
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Davide Stefanoni
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Alessandrini
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Degl'Innocenti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sanvito
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gregorio Maria Bergonzi
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Hematology and Stem Cell Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Annoni
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maddalena Panigada
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eleonora Cantoni
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniele Canarutto
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stephanie Z Xie
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Denver, CO, USA
| | - Raffaella Di Micco
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- University School of Advanced Studies IUSS, Pavia, Italy
| | - Alessandro Aiuti
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Hematology and Stem Cell Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giacomo De Luca
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Matucci-Cerinic
- Vita-Salute San Raffaele University, Milan, Italy
- Inflammation Fibrosis and Ageing Initiative (INFLAGE), Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- National Research Council, Institute for Biomedical Technologies, Segrate, Italy
| | - Simone Cenci
- Vita-Salute San Raffaele University, Milan, Italy
- Inflammation Fibrosis and Ageing Initiative (INFLAGE), Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serena Scala
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Translational Medicine, Novartis Pharma, Basel, Switzerland
| | - Luigi Naldini
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Samuele Ferrari
- Vita-Salute San Raffaele University, Milan, Italy.
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
25
|
Peters F, Höfs W, Lee H, Brodesser S, Kruse K, Drexler HC, Hu J, Raker VK, Lukas D, von Stebut E, Krönke M, Niessen CM, Wickström SA. Sphingolipid metabolism orchestrates establishment of the hair follicle stem cell compartment. J Cell Biol 2025; 224:e202403083. [PMID: 39879198 PMCID: PMC11778283 DOI: 10.1083/jcb.202403083] [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: 03/14/2024] [Revised: 11/04/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
Sphingolipids serve as building blocks of membranes to ensure subcellular compartmentalization and facilitate intercellular communication. How cell type-specific lipid compositions are achieved and what is their functional significance in tissue morphogenesis and maintenance has remained unclear. Here, we identify a stem cell-specific role for ceramide synthase 4 (CerS4) in orchestrating fate decisions in skin epidermis. Deletion of CerS4 prevents the proper development of the adult hair follicle bulge stem cell (HFSC) compartment due to altered differentiation trajectories. Mechanistically, HFSC differentiation defects arise from an imbalance of key ceramides and their derivate sphingolipids, resulting in hyperactivation of noncanonical Wnt signaling. This impaired HFSC compartment establishment leads to disruption of hair follicle architecture and skin barrier function, ultimately triggering a T helper cell 2-dominated immune infiltration resembling human atopic dermatitis. This work uncovers a fundamental role for a cell state-specific sphingolipid profile in stem cell homeostasis and in maintaining an intact skin barrier.
Collapse
Affiliation(s)
- Franziska Peters
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- Department Cell Biology of the Skin, Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Center for Molecular Medicine Cologne, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Windie Höfs
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Hunki Lee
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Susanne Brodesser
- Faculty of Medicine and University Hospital of Cologne, Cluster of Excellence on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne, Germany
| | - Kai Kruse
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | | | - Jiali Hu
- Department Cell Biology of the Skin, Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Center for Molecular Medicine Cologne, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Dermatology, University of Cologne, Cologne, Germany
| | - Verena K. Raker
- Department of Dermatology, University of Münster, Münster, Germany
| | - Dominika Lukas
- Department of Dermatology, University of Cologne, Cologne, Germany
| | | | - Martin Krönke
- Faculty of Medicine and University Hospital of Cologne, Cluster of Excellence on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Carien M. Niessen
- Department Cell Biology of the Skin, Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Center for Molecular Medicine Cologne, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Sara A. Wickström
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| |
Collapse
|
26
|
Zhou X, Zhou Z, Qin X, Cheng J, Fu Y, Wang Y, Wang J, Qin P, Zhang D. Multiomics Analysis Reveals Neuroblastoma Molecular Signature Predicting Risk Stratification and Tumor Microenvironment Differences. J Proteome Res 2025; 24:1606-1623. [PMID: 39762147 DOI: 10.1021/acs.jproteome.4c00882] [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] [Indexed: 04/05/2025]
Abstract
Neuroblastoma (NB) remains associated with high mortality and low initial response rate, especially for high-risk patients, thus warranting exploration of molecular markers for precision risk classifiers. Through integrating multiomics profiling, we identified a range of hub genes involved in cell cycle and associated with dismal prognosis and malignant cells. Single-cell transcriptome sequencing revealed that a subset of malignant cells, subcluster 1, characterized by high proliferation and dedifferentiation, was strongly correlated with the hub gene signature and orchestrated an immunosuppressive tumor microenvironment (TME). Furthermore, we constructed a robust malignant subcluster 1 related signature (MSRS), which was an independent prognostic factor and superior to other clinical characteristics and published signatures. Besides, TME differences conferred remarkably distinct therapeutic responses between high and low MSRS groups. Notably, polo-like kinase-1 (PLK1) was one of the most crucial contributors to MSRS and remarkably correlated with malignant subcluster 1, and PLK1 inhibition was effective for NB treatment as demonstrated by in silico analysis and in vitro experiments. Overall, our study constructs a novel molecular model to further guide the clinical classification and individualized treatment of NB.
Collapse
Affiliation(s)
- Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaohan Qin
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jian Cheng
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yongcheng Fu
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuanyuan Wang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jingyue Wang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Pan Qin
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Da Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
27
|
Merle C, Rodrigues C, Pourkhalili Langeroudi A, Journot R, Rost F, Dang Y, Rulands S, Fre S. Transcriptional landscapes underlying Notch-induced lineage conversion and plasticity of mammary basal cells. EMBO J 2025:10.1038/s44318-025-00424-1. [PMID: 40186028 DOI: 10.1038/s44318-025-00424-1] [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: 10/22/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 04/07/2025] Open
Abstract
The mammary epithelium derives from multipotent mammary stem cells (MaSCs) that engage into differentiation during embryonic development. However, adult MaSCs maintain the ability to reactivate multipotency in non-physiological contexts. We previously reported that Notch1 activation in committed basal cells triggers a basal-to-luminal cell fate switch in the mouse mammary gland. Here, we report conservation of this mechanism and found that in addition to the mammary gland, constitutive Notch1 signaling induces a basal-to-luminal cell fate switch in adult cells of the lacrimal gland, the salivary gland, and the prostate. Since the lineage transition is progressive in time, we performed single-cell transcriptomic analysis on index-sorted mammary cells at different stages of lineage conversion, generating a temporal map of changes in cell identity. Combining single-cell analyses with organoid assays, we demonstrate that cell proliferation is indispensable for this lineage conversion. We also reveal the individual transcriptional landscapes underlying the cellular plasticity switching of committed mammary cells in vivo with spatial and temporal resolution. Given the roles of Notch signaling in cancer, these results may help to better understand the mechanisms that drive cellular transformation.
Collapse
Affiliation(s)
- Candice Merle
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Calvin Rodrigues
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Atefeh Pourkhalili Langeroudi
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Robin Journot
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Fabian Rost
- Max-Planck-Institute for the Physics of Complex Systems, Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | - Yiteng Dang
- Max-Planck-Institute for the Physics of Complex Systems, Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
- Center for Systems Biology, Dresden, Germany
| | - Steffen Rulands
- Ludwig-Maximilians-Universität München, Arnold-Sommerfeld-Center for Theoretical Physics, München, Germany
| | - Silvia Fre
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France.
| |
Collapse
|
28
|
Chen ZH, Pan TB, Zhang YH, Wang B, Sun XL, Gao M, Sun Y, Xu M, Han S, Shi X, Correa-da-Silva F, Yang C, Guo J, Wu H, Li YZ, Liu XQ, Gao F, Xu Z, Xu S, Liu X, Zhu Y, Deng Z, Liu S, Zhou Y, Yi CX, Liu L, Wu QF. Transcriptional conservation and evolutionary divergence of cell types across mammalian hypothalamus development. Dev Cell 2025:S1534-5807(25)00156-X. [PMID: 40203835 DOI: 10.1016/j.devcel.2025.03.009] [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: 05/31/2024] [Revised: 02/07/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025]
Abstract
The hypothalamus, an "ancient" subcortical brain structure, maintains physiological homeostasis and controls native behaviors. The evolution of homeostatic regulation and behavioral control in mammals may rely on adaptable neuronal identity establishment but conserved neural patterning mechanisms during neurodevelopment. Here, we combined single-cell, single-nucleus, and spatial transcriptomic datasets to map the spatial patterning of diverse progenitor domains and reconstruct their neurogenic lineages in the developing human and mouse hypothalamus. While the regional organizers orchestrating neural patterning are conserved between primates and rodents, we identified a human-enriched neuronal subtype and found a substantial increase in neuromodulatory gene expression among human neurons. Furthermore, cross-species comparison demonstrated a potential redistribution of two neuroendocrine neuronal subtypes and a shift in inter-transmitter and transmitter-peptide coupling within hypothalamic dopamine neurons. Together, our study lays a critical foundation for understanding cellular development and evolution of the mammalian hypothalamus.
Collapse
Affiliation(s)
- Zhen-Hua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | | | - Yu-Hong Zhang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Ben Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Xue-Lian Sun
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | | | - Yang Sun
- BGI Research, Beijing 102601, China
| | - Mingrui Xu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | | | - Xiang Shi
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Felipe Correa-da-Silva
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | | | - Junfu Guo
- BGI Research, Beijing 102601, China; BGI Research, Shenzhen 518083, China
| | - Haoda Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Zheng Li
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiu-Qin Liu
- Department of Obstetrics and Gynecology, Baoding Second Central Hospital, Baoding 072750, China
| | - Fei Gao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhiheng Xu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Shengjin Xu
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xin Liu
- BGI Research, Beijing 102601, China
| | - Ying Zhu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University Shanghai, Shanghai 200032, China
| | | | | | - Yi Zhou
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | | | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China; Beijing Key Laboratory for Genetics of Birth Defects, Beijing 100045, China.
| |
Collapse
|
29
|
Meng X, Zhu Y, Liu K, Wang Y, Liu X, Liu C, Zeng Y, Wang S, Gao X, Shen X, Chen J, Tao S, Xu Q, Dong L, Shen L, Wang L. CXXC-finger protein 1 associates with FOXP3 to stabilize homeostasis and suppressive functions of regulatory T cells. eLife 2025; 13:RP103417. [PMID: 40183773 PMCID: PMC11970909 DOI: 10.7554/elife.103417] [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] [Indexed: 04/05/2025] Open
Abstract
FOXP3-expressing regulatory T (Treg) cells play a pivotal role in maintaining immune homeostasis and tolerance, with their activation being crucial for preventing various inflammatory responses. However, the mechanisms governing the epigenetic program in Treg cells during their dynamic activation remain unclear. In this study, we demonstrate that CXXC-finger protein 1 (CXXC1) interacts with the transcription factor FOXP3 and facilitates the regulation of target genes by modulating H3K4me3 deposition. Cxxc1 deletion in Treg cells leads to severe inflammatory disease and spontaneous T cell activation, with impaired immunosuppressive function. As a transcriptional regulator, CXXC1 promotes the expression of key Treg functional markers under steady-state conditions, which are essential for the maintenance of Treg cell homeostasis and their suppressive functions. Epigenetically, CXXC1 binds to the genomic regulatory regions of Treg program genes in mouse Treg cells, overlapping with FOXP3-binding sites. Given its critical role in Treg cell homeostasis, CXXC1 presents itself as a promising therapeutic target for autoimmune diseases.
Collapse
Affiliation(s)
- Xiaoyu Meng
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Yezhang Zhu
- Department of Hematology, Tongji Hospital, School of Medicine, Tongji UniversityShanghaiChina
- Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center of Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji UniversityShanghaiChina
| | - Kuai Liu
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Yuxi Wang
- Laboratory Animal Center, Zhejiang UniversityHangzhouChina
| | - Xiaoqian Liu
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Chenxin Liu
- Zhejiang University School of MedicineHangzhouChina
| | - Yan Zeng
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Shuai Wang
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Xianzhi Gao
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Xin Shen
- Co-Facility Center, Zhejiang University School of MedicineHangzhouChina
| | - Jing Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Sijue Tao
- Laboratory Animal Center, Zhejiang UniversityHangzhouChina
| | - Qianying Xu
- Zhejiang University School of MedicineHangzhouChina
| | - Linjia Dong
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical CollegeHangzhouChina
| | - Li Shen
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhouChina
- Department of Orthopedics Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Lie Wang
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
- Laboratory Animal Center, Zhejiang UniversityHangzhouChina
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang UniversityJiaxingChina
| |
Collapse
|
30
|
Liu X, Zhang L, Li X, Chen L, Lu L, Yang Y, Wu Y, Zheng L, Tang J, Wang F, Han Y, Song X, Cao W, Li T. Single-cell multi-omics profiling uncovers the immune heterogeneity in HIV-infected immunological non-responders. EBioMedicine 2025; 115:105667. [PMID: 40184908 PMCID: PMC12002939 DOI: 10.1016/j.ebiom.2025.105667] [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: 09/06/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Immunological non-responders (INRs) are people living with HIV-1 who fail to achieve full immune reconstitution despite long-term effective antiretroviral therapy (ART). This incomplete recovery of CD4+ T cells increase the risk of opportunistic infections and non-AIDS-related morbidity and mortality. Understanding the mechanisms driving this immune dysfunction is critical for developing targeted therapies. METHODS We performed single-cell RNA sequencing (scRNA-seq) and single-cell VDJ sequencing (scVDJ-seq) on peripheral blood mononuclear cells (PBMCs) from INRs, immune responders (IRs), and healthy controls (HCs). We developed scGeneANOVA, a novel mixed model differential gene analysis tool, to detect differentially expressed genes and pathways. In addition, we developed the Viral Identification and Load Detection Analysis (VILDA) tool to quantify HIV-1 transcripts and investigate their relationship with interferon (IFN) pathway activation. FINDINGS Our analysis revealed that INRs exhibit a dysregulated IFN response, closely associated with CD4+ T cell exhaustion and immune recovery failure. The scGeneANOVA tool identified critical genes and pathways that were missed by traditional analysis methods, while VILDA showed higher levels of HIV-1 transcripts in INRs, which may drive the heightened IFN response. These findings support a potential contribution of IFN signalling in INR-related immune dysfunction. INTERPRETATION Our study provides new insights into the pathogenic mechanisms behind immune recovery failure in INRs, suggesting that IFN signalling might be involved in the development of CD4+ T cell exhaustion. The identification of key genes and pathways offers potential biomarkers and therapeutic targets for improving immune recovery in this vulnerable population. FUNDING This study was supported by the grants from Special Research Fund for the Central High-level Hospitals of Peking Union Medical College Hospital (Grant No. 2022-PUMCH-D-008), Chinese Academy of Medical Sciences (CAMS) Innovation Fund for Medical Sciences (Grant No. 2021-I2M-1-037), National Key Technologies R&D Program for the 13th Five-year Plan (Grant No. 2017ZX10202101-001). The funders played no role in the design, experiment conduction, data analysis and preparation of the manuscript of this work.
Collapse
Affiliation(s)
- Xiaosheng Liu
- School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China; Centre for Life Sciences, Tsinghua University, 100084, Beijing, China; Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Leidan Zhang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Xiaodi Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Ling Chen
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Lianfeng Lu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Yang Yang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Yuanni Wu
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Liyuan Zheng
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Jia Tang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Fada Wang
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Yang Han
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Xiaojing Song
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Wei Cao
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China; State Key Laboratory for Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, 100730, Beijing, China; Center for AIDS Research, Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Taisheng Li
- School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China; Centre for Life Sciences, Tsinghua University, 100084, Beijing, China; Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 100730, Beijing, China; State Key Laboratory for Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, 100730, Beijing, China; Center for AIDS Research, Chinese Academy of Medical Sciences, 100730, Beijing, China.
| |
Collapse
|
31
|
Kitani A, Matsui Y. Integrative network analysis reveals novel moderators of Aβ-Tau interaction in Alzheimer's disease. Alzheimers Res Ther 2025; 17:70. [PMID: 40176187 PMCID: PMC11967117 DOI: 10.1186/s13195-025-01705-x] [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: 10/11/2024] [Accepted: 02/25/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Although interactions between amyloid-beta and tau proteins have been implicated in Alzheimer's disease (AD), the precise mechanisms by which these interactions contribute to disease progression are not yet fully understood. Moreover, despite the growing application of deep learning in various biomedical fields, its application in integrating networks to analyze disease mechanisms in AD research remains limited. In this study, we employed BIONIC, a deep learning-based network integration method, to integrate proteomics and protein-protein interaction data, with an aim to uncover factors that moderate the effects of the Aβ-tau interaction on mild cognitive impairment (MCI) and early-stage AD. METHODS Proteomic data from the ROSMAP cohort were integrated with protein-protein interaction (PPI) data using a Deep Learning-based model. Linear regression analysis was applied to histopathological and gene expression data, and mutual information was used to detect moderating factors. Statistical significance was determined using the Benjamini-Hochberg correction (p < 0.05). RESULTS Our results suggested that astrocytes and GPNMB + microglia moderate the Aβ-tau interaction. Based on linear regression with histopathological and gene expression data, GFAP and IBA1 levels and GPNMB gene expression positively contributed to the interaction of tau with Aβ in non-dementia cases, replicating the results of the network analysis. CONCLUSIONS These findings suggest that GPNMB + microglia moderate the Aβ-tau interaction in early AD and therefore are a novel therapeutic target. To facilitate further research, we have made the integrated network available as a visualization tool for the scientific community (URL: https://igcore.cloud/GerOmics/AlzPPMap ).
Collapse
Affiliation(s)
- Akihiro Kitani
- Department of Integrated Health Science, Biomedical and Health Informatics Unit, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yusuke Matsui
- Department of Integrated Health Science, Biomedical and Health Informatics Unit, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Institute for Glyco-Core Research (Igcore), Nagoya University, Nagoya, Aichi, 461-8673, Japan.
| |
Collapse
|
32
|
Ma H, Srivastava S, Ho SWT, Xu C, Lian BSX, Ong X, Tay ST, Sheng T, Lum HYJ, Abdul Ghani SAB, Chu Y, Huang KK, Goh YT, Lee M, Hagihara T, Ng CSY, Tan ALK, Zhang Y, Ding Z, Zhu F, Ng MSW, Joseph CRC, Chen H, Li Z, Zhao JJ, Rha SY, Teh M, Yeong J, Yong WP, So JBY, Sundar R, Tan P. Spatially Resolved Tumor Ecosystems and Cell States in Gastric Adenocarcinoma Progression and Evolution. Cancer Discov 2025; 15:767-792. [PMID: 39774838 PMCID: PMC11962405 DOI: 10.1158/2159-8290.cd-24-0605] [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: 05/03/2024] [Revised: 10/17/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025]
Abstract
SIGNIFICANCE Integration of spatial transcriptomic (GeoMx Digital Spatial Profiler) and single-cell RNA sequencing data from multiple gastric cancers identifies spatially resolved expression-based intratumoral heterogeneity, associated with distinct immune microenvironments. We uncovered two separate evolutionary trajectories associated with specific molecular subtypes, clinical prognoses, stromal neighborhoods, and genetic drivers. Tumor-stroma interfaces emerged as a unique state of tumor ecology.
Collapse
Affiliation(s)
- Haoran Ma
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Supriya Srivastava
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shamaine Wei Ting Ho
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Chang Xu
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | | | - Xuewen Ong
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Su Ting Tay
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Taotao Sheng
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | | | | | - Yunqiang Chu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Kie Kyon Huang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Yeek Teck Goh
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Minghui Lee
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Takeshi Hagihara
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Clara Shi Ya Ng
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Angie Lay Keng Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Yanrong Zhang
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore, Singapore
| | - Zichen Ding
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Zhu
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michelle Shu Wen Ng
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Craig Ryan Cecil Joseph
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Hui Chen
- MGI Tech Singapore Pte. Ltd., Singapore, Singapore
| | - Zhen Li
- MGI Tech Singapore Pte. Ltd., Singapore, Singapore
| | - Joseph J. Zhao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ming Teh
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Joe Yeong
- Department of Pathology, National University Hospital, Singapore, Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Wei Peng Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
| | - Jimmy Bok-Yan So
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore, Singapore
- Division of Surgical Oncology, National University Cancer Institute, Singapore, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raghav Sundar
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
| | - Patrick Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore
- Singhealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, Singapore
| |
Collapse
|
33
|
Norton EG, Chapman NM, Shi H, Meng X, Huang H, KC A, Rankin S, Saravia J, Yuan S, Hu H, Vogel P, Chi H. Vps34-orchestrated lipid signaling processes regulate the transitional heterogeneity and functional adaptation of effector regulatory T cells. PLoS Biol 2025; 23:e3003074. [PMID: 40215232 PMCID: PMC11990774 DOI: 10.1371/journal.pbio.3003074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/19/2025] [Indexed: 04/14/2025] Open
Abstract
Regulatory T cell (Treg) heterogeneity exists in lymphoid and non-lymphoid tissues, but we have limited understanding of context-dependent functions and spatiotemporal regulators of heterogenous Treg states, especially during perinatal life when immune tolerance is established. Here, we revealed that the class III PI3K Vps34 orchestrates effector Treg (eTreg) transitional heterogeneity during perinatal life. We found that loss of Vps34 reduced terminal eTreg accumulation in lymphoid tissues, associated with decreased Treg generation in non-lymphoid tissues and development of an early-onset autoimmune-like disease. After perinatal life, Vps34-deficient eTreg accumulation was further impaired due to reduced cell survival, highlighting temporal regulation of eTreg heterogeneity and maintenance by Vps34. Accordingly, inhibition of Vps34 in mature Tregs disrupted immune homeostasis but boosted anti-tumor immunity. Mechanistically, multiomics profiling approaches uncovered that Vps34-orchestrated transcriptional and epigenetic remodeling promotes terminal eTreg programming. Further, via genetic deletion of the Vps34-interacting proteins Atg14 or Uvrag in Tregs, we established that Atg14 but not Uvrag was required for the overall survival, but not terminal differentiation, of eTregs, suggesting that autophagy but not endocytosis partly contributed to Vps34-dependent effects. Accordingly, mice with Treg-specific loss of Atg14, but not Uvrag, had moderately disrupted immune homeostasis and reduced tumor growth, with Vps34- or Atg14-dependent gene signatures also being elevated in intratumoral Tregs from human cancer patients. Collectively, our study reveals distinct Vps34-orchestrated signaling events that regulate eTreg heterogeneity and functional adaptation and the pathophysiological consequences on autoimmunity versus anti-tumor immunity.
Collapse
Affiliation(s)
- Erienne G. Norton
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Nicole M. Chapman
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Hao Shi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Xiaoxi Meng
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Hongling Huang
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Anil KC
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Sherri Rankin
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Jordy Saravia
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Sujing Yuan
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Haoran Hu
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Peter Vogel
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Hongbo Chi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
34
|
Li Y, Hu M, Zhang Z, Wu B, Zheng J, Zhang F, Hao J, Xue T, Li Z, Zhu C, Liu Y, Zhao L, Xu W, Xin P, Feng C, Wang W, Zhao Y, Qiu Q, Wang K. Origin and stepwise evolution of vertebrate lungs. Nat Ecol Evol 2025; 9:672-691. [PMID: 39953253 DOI: 10.1038/s41559-025-02642-6] [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: 10/20/2023] [Accepted: 01/15/2025] [Indexed: 02/17/2025]
Abstract
Lungs are essential respiratory organs in terrestrial vertebrates, present in most bony fishes but absent in cartilaginous fishes, making them an ideal model for studying organ evolution. Here we analysed single-cell RNA sequencing data from adult and developing lungs across vertebrate species, revealing significant similarities in cell composition, developmental trajectories and gene expression patterns. Surprisingly, a large proportion of lung-related genes, coexpression patterns and many lung enhancers are present in cartilaginous fishes despite their lack of lungs, suggesting that a substantial genetic foundation for lung development existed in the last common ancestor of jawed vertebrates. In addition, the 1,040 enhancers that emerged since the last common ancestor of bony fishes probably contain lung-specific elements that led to the development of lungs. We further identified alveolar type 1 cells as a mammal-specific alveolar cell type, along with several mammal-specific genes, including ager and sfta2, that are highly expressed in lungs. Functional validation showed that deletion of sfta2 in mice leads to severe respiratory defects, highlighting its critical role in mammalian lung features. Our study provides comprehensive insights into the evolution of vertebrate lungs, demonstrating how both regulatory network modifications and the emergence of new genes have shaped lung development and specialization across species.
Collapse
Affiliation(s)
- Ye Li
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Mingliang Hu
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Zhigang Zhang
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Baosheng Wu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jiangmin Zheng
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Fenghua Zhang
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Jiaqi Hao
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Tingfeng Xue
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Zhaohong Li
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Chenglong Zhu
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Yuxuan Liu
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Lei Zhao
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Wenjie Xu
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Peidong Xin
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Chenguang Feng
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China.
| | - Wen Wang
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China.
- New Cornerstone Science Laboratory, Xi'an, China.
| | - Yilin Zhao
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China.
| | - Qiang Qiu
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China.
| | - Kun Wang
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| |
Collapse
|
35
|
He W, Luo Q, Zhao J, Wang M, Zhao A, Feng L, Reda A, Lindgren E, Stukenborg J, Chen J, Deng Q. X-Linked Gene Dosage and SOX2 Act as Key Roadblocks for Human Germ Cell Specification in Klinefelter Syndrome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410533. [PMID: 39996497 PMCID: PMC12005746 DOI: 10.1002/advs.202410533] [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: 08/30/2024] [Revised: 02/03/2025] [Indexed: 02/26/2025]
Abstract
Klinefelter syndrome (KS), characterized by the presence of at least one extra X-chromosome, is a common cause of male infertility. However, the mechanism underlying the failure of germline specification is not well studied. Intriguingly, the differentiation efficiency of female human pluripotent stem cells (hPSCs) is often lower than that of male. This study investigates how X-linked gene dosage affects human primordial germ cell-like cells (hPGCLCs) specification in both healthy and diseased conditions. This work reveals that X-linked genes play a multifaceted role against the fate competency to hPGCLCs, with escape genes IGSF1 and CHRDL1 inhibiting the TGF-beta/Activin A and BMP pathways, respectively. Notably, this work identifies a previously unrecognized role of SOX2, upregulated by the escape gene USP9X, elucidating a species-specific function in the mammalian germline. The USP9X-SOX2 regulatory axis profoundly influenced cellular metabolism, mitochondrial morphology, and progenitor competence in hPGCLCs specification. Furthermore, the inability to downregulate SOX2 and upregulate SOX17 in response to BMP signaling impedes downstream gene activation due to motif binding competition. These findings shed novel insights into the human germline specification by elucidating the divergent roles of SOX2 versus SOX17 in mammals, influenced by X-linked gene dosage effects. These results offer potential applications for improving the induction efficiency of hPGCLCs, facilitating disease mechanistic studies.
Collapse
Affiliation(s)
- Wenteng He
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Qing Luo
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Jian Zhao
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
- Department of Oncology‐PathologyKarolinska InstitutetStockholm171 77Sweden
| | - Mengting Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Allan Zhao
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Luohua Feng
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Ahmed Reda
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Eva Lindgren
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
| | - Jan‐Bernd Stukenborg
- NORDFERTIL Research Lab StockholmChildhood Cancer Research UnitDepartment of Women's and Children's HealthKarolinska InstitutetKarolinska University HospitalStockholm17 165Sweden
| | - Jiayu Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
- Frontier Science Center for Stem Cell ResearchTongji UniversityShanghai200092China
| | - Qiaolin Deng
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm171 77Sweden
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UnviersityStockholm11418Sweden
| |
Collapse
|
36
|
Ravi K, Zhang Y, Sakala L, Manoharan TJM, Pockaj B, LaBaer J, Park JG, Nikkhah M. Tumor Microenvironment On-A-Chip and Single-Cell Analysis Reveal Synergistic Stromal-Immune Crosstalk on Breast Cancer Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413457. [PMID: 40056038 PMCID: PMC12021108 DOI: 10.1002/advs.202413457] [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: 10/22/2024] [Revised: 02/11/2025] [Indexed: 04/26/2025]
Abstract
Solid tumors develop within a complex environment called the tumor microenvironment (TME), which is sculpted by the presence of other cells, such as cancer-associated fibroblasts (CAFs) and immune cells like macrophages (Mφs). Despite the presence of immune cells, tumor cells orchestrate a tumor-supportive environment through intricate interaction with the components of the TME. However, the specific mechanism by which this intercellular dialogue is regulated is not fully understood. To that end, the development of an organotypic 3D breast TME-on-a-chip (TMEC) model, integrated with single-cell RNA sequencing analysis, is reported to mechanistically evaluate the progression of triple-negative breast cancer (TNBC) cells in the presence of patient-derived CAFs and Mφs. Extensive functional assays, including invasion and morphometric characterization, reveal the synergistic influence of CAFs and Mφs on tumor cells. Furthermore, gene expression and pathway enrichment analyses identify the involvement of the KYNU gene, suggesting a potential immune evasion mechanism through the kynurenine pathway. Lastly, the pharmacological targeting of the identified pathway is investigated.
Collapse
Affiliation(s)
- Kalpana Ravi
- School of Biological and Health Systems Engineering (SBHSE)Arizona State UniversityTempeAZ85287USA
| | - Yining Zhang
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | - Lydia Sakala
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | | | | | - Joshua LaBaer
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | - Jin G. Park
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE)Arizona State UniversityTempeAZ85287USA
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| |
Collapse
|
37
|
Robinson MA, Kung SHY, Youssef KYM, Scheck KM, Bell RH, Sar F, Haegert AM, Asmae MM, Cheng C, Yeack SV, Mathur BT, Jiang F, Collins CC, Hach F, Willerth SM, Flannigan RK. 3D Bioprinted Coaxial Testis Model Using Human Induced Pluripotent Stem Cells:A Step Toward Bicompartmental Cytoarchitecture and Functionalization. Adv Healthc Mater 2025; 14:e2402606. [PMID: 39955738 PMCID: PMC12004438 DOI: 10.1002/adhm.202402606] [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: 07/15/2024] [Revised: 02/04/2025] [Indexed: 02/17/2025]
Abstract
Fertility preservation following pediatric cancer therapy programs has become a major avenue of infertility research. In vitro spermatogenesis (IVS) aims to generate sperm from banked prepubertal testicular tissues in a lab setting using specialized culture conditions. While successful using rodent tissues, progress with human tissues is limited by the scarcity of human prepubertal testicular tissues for research. This study posits that human induced pluripotent stem cells (hiPSCs) can model human prepubertal testicular tissue to facilitate the development of human IVS conditions. Testicular cells derived from hiPSCs are characterized for phenotype markers and profiled transcriptionally. HiPSC-derived testicular cells are bioprinted into core-shell constructs representative of testis cytoarchitecture and found to capture functional aspects of prepubertal testicular tissues within 7 days under xeno-free conditions. Moreover, hiPSC-derived Sertoli cells illustrate the capacity to mature under pubertal-like conditions. The utility of the model is tested by comparing 2 methods of supplementing retinoic acid (RA), the vitamin responsible for inducing spermatogenesis. The model reveals a significant gain in activity under microsphere-released RA compared to RA medium supplementation, indicating that the fragility of free RA in vitro may be a contributing factor to the molecular dysfunction observed in human IVS studies to date.
Collapse
Affiliation(s)
| | - Sonia HY Kung
- Vancouver Prostate CentreVancouverBritish ColumbiaV6H 3Z6Canada
| | | | - Kali M Scheck
- Axolotl BiosciencesVictoriaBritish ColumbiaV8W 2Y2Canada
| | - Robert H Bell
- Vancouver Prostate CentreVancouverBritish ColumbiaV6H 3Z6Canada
| | - Funda Sar
- Vancouver Prostate CentreVancouverBritish ColumbiaV6H 3Z6Canada
| | - Anne M Haegert
- Vancouver Prostate CentreVancouverBritish ColumbiaV6H 3Z6Canada
| | - M Mahdi Asmae
- Vancouver Prostate CentreVancouverBritish ColumbiaV6H 3Z6Canada
| | - Changfeng Cheng
- Faculty of ForestryUniversity of British ColumbiaVancouverBritish ColumbiaV6T 1Z4Canada
| | - Salina V Yeack
- Axolotl BiosciencesVictoriaBritish ColumbiaV8W 2Y2Canada
| | - Bhairvi T Mathur
- Faculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaV6T 1Z4Canada
| | - Feng Jiang
- Faculty of ForestryUniversity of British ColumbiaVancouverBritish ColumbiaV6T 1Z4Canada
| | - Colin C Collins
- Vancouver Prostate CentreVancouverBritish ColumbiaV6H 3Z6Canada
| | - Faraz Hach
- Vancouver Prostate CentreVancouverBritish ColumbiaV6H 3Z6Canada
| | - Stephanie M Willerth
- Faculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaV6T 1Z4Canada
- Department of Mechanical EngineeringUniversity of VictoriaVictoriaBritish ColumbiaV8P 5C2Canada
- Division of Medical SciencesUniversity of VictoriaVictoriaBritish ColumbiaV8P 5C2Canada
| | - Ryan K Flannigan
- Vancouver Prostate CentreVancouverBritish ColumbiaV6H 3Z6Canada
- Department of Urologic SciencesUniversity of British ColumbiaVancouverBritish ColumbiaV6T 1Z4Canada
| |
Collapse
|
38
|
Bhattacharya S, Tie G, Singh PNP, Malagola E, Eskiocak O, He R, Kraiczy J, Gu W, Perlov Y, Alici-Garipcan A, Beyaz S, Wang TC, Zhou Q, Shivdasani RA. Intestinal secretory differentiation reflects niche-driven phenotypic and epigenetic plasticity of a common signal-responsive terminal cell. Cell Stem Cell 2025:S1934-5909(25)00095-5. [PMID: 40203837 DOI: 10.1016/j.stem.2025.03.005] [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: 09/19/2024] [Revised: 12/27/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025]
Abstract
Enterocytes and four classic secretory cell types derive from intestinal epithelial stem cells. Based on morphology, location, and canonical markers, goblet and Paneth cells are considered distinct secretory types. Here, we report high overlap in their transcripts and sites of accessible chromatin, in marked contrast to those of their enteroendocrine or tuft cell siblings. Mouse and human goblet and Paneth cells express extraordinary fractions of few antimicrobial genes, which reflect specific responses to local niches. Wnt signaling retains some ATOH1+ secretory cells in crypt bottoms, where the absence of BMP signaling potently induces Paneth features. Cells that migrate away from crypt bottoms encounter BMPs and thereby acquire goblet properties. These phenotypes and underlying accessible cis-elements interconvert in post-mitotic cells. Thus, goblet and Paneth properties represent alternative phenotypic manifestations of a common signal-responsive terminal cell type. These findings reveal exquisite niche-dependent cell plasticity and cis-regulatory dynamics in likely response to antimicrobial needs.
Collapse
Affiliation(s)
- Swarnabh Bhattacharya
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Guodong Tie
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Pratik N P Singh
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Onur Eskiocak
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Graduate Program in Genetics, State University of New York, Stony Brook, NY 11794, USA
| | - Ruiyang He
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Judith Kraiczy
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Gu
- Division of Regenerative Medicine & Hartman Institute for Therapeutic Organ Regeneration, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yakov Perlov
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Qiao Zhou
- Division of Regenerative Medicine & Hartman Institute for Therapeutic Organ Regeneration, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
39
|
Tang Z, Bai Y, Fang Q, Yuan Y, Zeng Q, Chen S, Xu T, Chen J, Tan L, Wang C, Li Q, Lin J, Yang Z, Wu X, Shi G, Wang J, Yin C, Guo J, Liu S, Peng S, Kuang M. Spatial transcriptomics reveals tryptophan metabolism restricting maturation of intratumoral tertiary lymphoid structures. Cancer Cell 2025:S1535-6108(25)00112-6. [PMID: 40185093 DOI: 10.1016/j.ccell.2025.03.011] [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: 06/27/2024] [Revised: 01/22/2025] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
Tertiary lymphoid structures (TLSs) are ectopic lymphoid aggregates found in numerous cancers, often linked to enhanced immunotherapy responses and better clinical outcomes. However, the factors driving TLS maturation are not fully understood. Using near single-cell spatial transcriptomic mapping, we comprehensively profile TLSs under various maturation stages and their microenvironment in hepatocellular carcinoma (HCC). Based on their developmental trajectories, we classify immature TLSs into two groups: conforming and deviating TLSs. Our findings indicate that conforming TLSs, similar to mature TLSs, possess a niche function for immunotherapy responses, while deviating TLSs do not. We discover that the tryptophan-enriched metabolic microenvironment shaped by malignant cells contributes to the deviation of TLS maturation. Inhibiting tryptophan metabolism promotes intratumoral TLS maturation and enhances tumor control, synergizing with anti-PD-1 treatments. Therefore, promoting TLS maturation represents a potential strategy to improve antitumor responses and immunotherapy outcomes.
Collapse
Affiliation(s)
- Zhonghui Tang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Yinqi Bai
- BGI Research, Sanya 572025, China; BGI Research, Hangzhou 310030, China
| | - Qi Fang
- BGI Research, Hangzhou 310030, China
| | - Yuchen Yuan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Qianwen Zeng
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Shuling Chen
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Tianyi Xu
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Jianyu Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Department of Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Li Tan
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Chunqing Wang
- BGI Research, Chongqing 401329, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Li
- BGI Research, Sanya 572025, China; BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinpei Lin
- BGI Research, Sanya 572025, China; BGI Research, Hangzhou 310030, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Zhuoxuan Yang
- BGI Research, Sanya 572025, China; BGI Research, Hangzhou 310030, China
| | - Xia Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Guowei Shi
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ji Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Changjun Yin
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Jianping Guo
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China; Shenzhen Bay Laboratory, Shenzhen 518000, China; Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518120, China; The Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou 510000, China.
| | - Sui Peng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Clinical Trial Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| | - Ming Kuang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
40
|
Llera-Oyola J, Pérez-Moraga R, Parras M, Rosón B. How to view the female reproductive tract through single-cell looking glasses. Am J Obstet Gynecol 2025; 232:S21-S43. [PMID: 40253081 DOI: 10.1016/j.ajog.2024.08.040] [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: 08/29/2023] [Revised: 07/04/2024] [Accepted: 08/24/2024] [Indexed: 04/21/2025]
Abstract
Single-cell technologies have emerged as an unprecedented tool for biologists and clinicians, allowing them to assess organs and tissues at the level of individual cells. In the field of women's reproductive biology, single-cell studies have provided insights into the cellular and molecular processes that regulate reproductive and obstetrical functions in health and disease. The knowledge that these studies generate is helping clinicians to improve the understanding and diagnosis of infertility related issues or pregnancy complications and to find new avenues for their treatment. However, navigating the expansive landscape of this type of transcriptomic data analysis represents a pivotal challenge in current research. Single cell RNA sequencing involves isolating cells into droplets, reverse transcribing RNA to generate complementary DNA, with each droplet content uniquely labeled by a barcode. Upon sequencing the complementary DNAs, the barcodes enable the reassignment of sequencing reads to individual droplets, facilitating the reconstruction of the cellular landscape of the sample obtained from a tissue or organ and beyond. Researchers, equipped with the metaphorical 'single-cell glasses,' must adequately choose from a plethora of strategies to dissect and interpret cellular information. Sophisticated algorithms and the decision-making process are often underestimated, resulting in artefactual or cumbersome interpreted results. Computational biologists apply and innovate computational tools designed to process, model, and interpret expansive datasets. The ramifications of their work extend far beyond the realm of data processing; they give shape to the outcome of analyses, playing a pivotal role in drawing meaningful conclusions from the wealth of information garnered. In this review, we describe the wide variety of approaches and analytical steps available with enough detail to gain a concise picture of what a complete examination of a single-cell dataset would be. We commence with a discussion on key points in experimental design, highlighting crucial questions one should consider. Following this, we delve into the various preprocessing and quality control steps essential for any single-cell dataset. The subsequent section offers a detailed guide on constructing a single-cell atlas, exploring nuances such as differential characteristics in visualization and clustering techniques, as well as strategies for assigning identity to cell populations through gene marker annotations. Moving beyond the creation of an atlas, we explore methods for investigating pathological conditions. This involves conducting cell population comparison tests between conditions and analyzing specific cell-to-cell communications and cellular differentiation trajectories in both health and disease scenarios. This work aims to furnish a newcomer researcher and/or clinician with essential guidelines to embark on a single-cell adventure without succumbing to common pitfalls. By bridging the gap between theory and practice, it facilitates the translation of single-cell technologies into clinically relevant applications. Throughout the manuscript, practical examples of its usage in women's reproductive health studies are provided. Various sections delve into specific clinical scenarios, demonstrating how these guidelines can be instrumental in unraveling the molecular landscapes of diseases and physiological processes related to women's reproduction.
Collapse
Affiliation(s)
- Jaime Llera-Oyola
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain
| | - Raúl Pérez-Moraga
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain; R&D Department, Igenomix, Valencia, Spain
| | - Marcos Parras
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain
| | - Beatriz Rosón
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain.
| |
Collapse
|
41
|
Lee C, Kim JE, Cha YE, Moon JH, Kim ER, Chang DK, Kim YH, Hong SN. IFN-γ-Induced intestinal epithelial cell-type-specific programmed cell death: PANoptosis and its modulation in Crohn's disease. Front Immunol 2025; 16:1523984. [PMID: 40230837 PMCID: PMC11994596 DOI: 10.3389/fimmu.2025.1523984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/04/2025] [Indexed: 04/16/2025] Open
Abstract
Background Crohn's disease (CD) is a chronic inflammatory bowel disease (IBD) and is considered a Th1-mediated disease, supported by the over-expression of interferon-gamma (IFN-γ) in the intestinal lamina propria. IFN-γ has a pleiotropic effect on the intestinal epithelial cells (IECs), suggesting that IFN-γ-induced responses may differ between epithelial cell types. Methods We established human small intestinal organoids (enteroids) derived from non-IBD controls and CD patients. Using human enteroids, the major response of IECs induced by IFN-γ was evaluated, focusing on the IFN-γ-induced programmed cell death (PCD) pathway. Identified IFN-γ-induced responses were validated in surgically resected intestinal samples and publicly available single-cell RNA-sequencing datasets. Results IFN-γ stimulated programmed cell death (PCD) of IECs in both control and CD enteroids in a dose-dependent manner. Pyroptosis, apoptosis. and necroptosis were activated in enteroids, suggesting that PANoptosis was the main process of IFN-γ-induced PCD in IECs. The response to IFN-γ depends on the cell type of the IECs. IFN-γ induced depletion of enterocytes with upregulation of PANoptosis-associated genes, while leading to expansion of goblet cells without significant change in PANoptosis-associated gene expression. Individual PCD inhibitors were insufficient to block IFN-γ-induced cytotoxicity, whereas the selective JAK1 inhibitor (upadacitinib) effectively blocked IFN-γ-induced cytotoxicity and PANoptosis. Furthermore, PANoptosis was significantly activated in surgically resected tissues and in publicly available single-cell RNA-sequencing datasets of intestinal tissues from patients with CD. Conclusion IFN-γ induces PANoptosis in enterocytes, which can be treated with a selective JAK1 inhibitor in patients with CD.
Collapse
Affiliation(s)
- Chansu Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Stem Cell and Regenerative Medicine Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Ji Eun Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yeo-Eun Cha
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Stem Cell and Regenerative Medicine Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Ji Hwan Moon
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Eun Ran Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong Kyung Chang
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young-Ho Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung Noh Hong
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Stem Cell and Regenerative Medicine Center, Samsung Medical Center, Seoul, Republic of Korea
| |
Collapse
|
42
|
Xu QZ, Li YX, Shi WG, Dong Y, Li Z, Ip JCH, Galaska MP, Han C, Zhang Q, Sun YY, Zhao LL, Sun KM, Wang ZL, Qiu JW, Zhang XL. Chromosome-level genome assembly and single-cell analysis unveil molecular mechanisms of arm regeneration in the ophiuroid Ophiura sarsii vadicola. Genome Biol 2025; 26:82. [PMID: 40165295 PMCID: PMC11959729 DOI: 10.1186/s13059-025-03542-5] [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: 07/09/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Ophiuroids, belonging to Ophiuroidea in Echinodermata, possess remarkable regenerative capacities in their arms, relying on cellular recruitment and de-differentiation. However, limited high-quality genomic resources have hindered the investigation of the underlying molecular mechanisms of ophiuroid regeneration. RESULTS Here, we report a chromosome-level genome of Ophiura sarsii vadicola, 259.28 Mbp in length with a scaffold N50 length of 66.91 Mbp. We then perform bulk and single-cell RNA sequencing analysis to investigate gene expression and cellular dynamics during arm regeneration. We identify five distinct cellular clusters involved in the arm regeneration and infer the dynamic transformations from sensory stimulation to injury response, wound healing, and tissue regeneration. We find that progenitor cells derived from connective tissue cells differentiate into muscle, cartilage, endothelial, and epithelial cells. Pseudotime analysis indicates that muscle differentiation occurs early in the regeneration process. CONCLUSIONS Our genomic resource and single-cell atlas shed light on the mechanisms of organ regeneration in ophiuroids.
Collapse
Affiliation(s)
- Qin-Zeng Xu
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, PR China.
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, PR China.
| | - Yi-Xuan Li
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, PR China
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, PR China
| | - Wen-Ge Shi
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, PR China
| | - Yue Dong
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, PR China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, PR China
| | - Zhong Li
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, PR China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, PR China
| | - Jack Chi-Ho Ip
- Science Unit, Lingnan University, Hong Kong SAR, PR China
| | - Matthew P Galaska
- Pacific Marine Environmental Lab, National Oceanic and Atmospheric Administration, Seattle, WA, USA
- Anchor QEA, Seattle, WA, USA
| | - Chen Han
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, PR China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, PR China
| | - Qian Zhang
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, PR China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, PR China
| | - Yu-Yao Sun
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, PR China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, PR China
| | - Lin-Lin Zhao
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, PR China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, PR China
| | - Kai-Ming Sun
- Institute of Oceanographic Instrumentation, Qilu University of Technology (Shandong Academy of Sciences), Qingdao, PR China
| | - Zong-Ling Wang
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, PR China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, PR China
| | - Jian-Wen Qiu
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, PR China
| | - Xue-Lei Zhang
- MNR Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, PR China
- Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, PR China
| |
Collapse
|
43
|
Hu X, Li H, Chen M, Qian J, Jiang H. Reference-informed evaluation of batch correction for single-cell omics data with overcorrection awareness. Commun Biol 2025; 8:521. [PMID: 40158033 PMCID: PMC11954866 DOI: 10.1038/s42003-025-07947-7] [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: 10/06/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
Batch effect correction (BEC) is fundamental to integrate multiple single-cell RNA sequencing datasets, and its success is critical to empower in-depth interrogation for biological insights. However, no simple metric is available to evaluate BEC performance with sensitivity to data overcorrection, which erases true biological variations and leads to false biological discoveries. Here, we propose RBET, a reference-informed statistical framework for evaluating the success of BEC. Using extensive simulations and six real data examples including scRNA-seq and scATAC-seq datasets with different numbers of batches, batch effect sizes and numbers of cell types, we demonstrate that RBET evaluates the performance of BEC methods more fairly with biologically meaningful insights from data, while other methods may lead to false results. Moreover, RBET is computationally efficient, sensitive to overcorrection and robust to large batch effect sizes. Thus, RBET provides a robust guideline on selecting case-specific BEC method, and the concept of RBET is extendable to other modalities.
Collapse
Affiliation(s)
- Xiaoyue Hu
- Center for Data Science, Zhejiang University, Hangzhou, China
- School of Mathematical Sciences, Zhejiang University, Hangzhou, China
| | - He Li
- Center for Data Science, Zhejiang University, Hangzhou, China
| | - Ming Chen
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Junbin Qian
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, China.
| | - Hangjin Jiang
- Center for Data Science, Zhejiang University, Hangzhou, China.
| |
Collapse
|
44
|
Liu J, Wang Y, Li C, Gu Y, Ono N, Welch J. CytoSimplex: visualizing single-cell fates and transitions on a simplex. Bioinformatics 2025; 41:btaf119. [PMID: 40119904 PMCID: PMC11992338 DOI: 10.1093/bioinformatics/btaf119] [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: 12/10/2023] [Revised: 01/23/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025] Open
Abstract
SUMMARY Cells differentiate to their final fates along unique trajectories, often involving multi-potent progenitors that can produce multiple terminally differentiated cell types. Recent developments in single-cell transcriptomic and epigenomic measurement provide tremendous opportunities for mapping these trajectories. The visualization of single-cell data often relies on dimension reduction methods such as UMAP to simplify high-dimensional single-cell data down to an understandable 2D form. However, these dimension reduction methods are not constructed to allow direct interpretation of the reduced dimensions in terms of cell differentiation. To address these limitations, we developed a new approach that places each cell from a single-cell dataset within a simplex whose vertices correspond to terminally differentiated cell types. Our approach can quantify and visualize current cell fate commitment and future cell potential. We developed CytoSimplex, a standalone open-source package implemented in R and Python that provides simple and intuitive visualizations of cell differentiation in 2D ternary and 3D quaternary plots. We believe that CytoSimplex can help researchers gain a better understanding of cell type transitions in specific tissues and characterize developmental processes. AVAILABILITY AND IMPLEMENTATION The R version of CytoSimplex is available on Github at https://github.com/welch-lab/CytoSimplex. The Python version of CytoSimplex is available on Github at https://github.com/welch-lab/pyCytoSimplex.
Collapse
Affiliation(s)
- Jialin Liu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Yichen Wang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Chen Li
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Yichen Gu
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, 77030, United States
| | - Joshua Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, United States
- Department of Computer Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| |
Collapse
|
45
|
Iliopoulou L, Tzaferis C, Prados A, Roumelioti F, Koliaraki V, Kollias G. Different fibroblast subtypes propel spatially defined ileal inflammation through TNFR1 signalling in murine ileitis. Nat Commun 2025; 16:3023. [PMID: 40155385 PMCID: PMC11953319 DOI: 10.1038/s41467-025-57570-7] [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: 03/05/2024] [Accepted: 02/25/2025] [Indexed: 04/01/2025] Open
Abstract
Crohn's disease (CD) is a persistent inflammatory disorder primarily affecting the terminal ileum. The TnfΔΑRE mice, which spontaneously develop CD-like ileitis due to TNF overexpression, represent a faithful model of the human disease. Here, via single-cell RNA sequencing in TnfΔΑRE mice, we show that murine TNF-dependent ileitis is characterized by cell expansion in tertiary lymphoid organs (TLO), T cell effector reprogramming, and accumulation of activated macrophages in the submucosal granulomas. Within the stromal cell compartment, fibroblast subsets (telocytes, trophocytes, PdgfraloCd81- cells) are less abundant while lymphatic endothelial cells (LEC) and fibroblastic reticular cells (FRC) show relative expansion compared to the wild type. All three fibroblast subsets show strong pro-inflammatory signature. TNFR1 loss or gain of function experiments in specific fibroblast subsets suggest that the TnfΔΑRE-induced ileitis is initiated in the lamina propria via TNF pathway activation in villus-associated fibroblasts (telocytes and PdgfraloCd81- cells), which are responsible for the organization of TLOs. Trophocytes drive disease progression in the submucosal layer, accompanied by the excessive formation of granulomas. These findings provide evidence for spatial regulation of inflammation by fibroblast subsets and underscore the pivotal role of fibroblasts in the inception and advancement of ileitis.
Collapse
Affiliation(s)
- Lida Iliopoulou
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Christos Tzaferis
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Alejandro Prados
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Fani Roumelioti
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Vasiliki Koliaraki
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - George Kollias
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
46
|
Lin Z, Wang W, Liu R, Li Q, Lee J, Hirschler C, Liu J. Cyborg organoids integrated with stretchable nanoelectronics can be functionally mapped during development. Nat Protoc 2025:10.1038/s41596-025-01147-7. [PMID: 40140634 DOI: 10.1038/s41596-025-01147-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/31/2024] [Indexed: 03/28/2025]
Abstract
Organoids are in vitro miniaturized cellular models of organs that offer opportunities for studying organ development, disease mechanisms and drug screening. Understanding the complex processes governing organoid development and function requires methods suitable for the continuous, long-term monitoring of cell activities (for example, electrophysiological and mechanical activity) at single-cell resolution throughout the entire three-dimensional (3D) structure. Cyborg organoid technology addresses this need by seamlessly integrating stretchable mesh nanoelectronics with tissue-like properties, such as tissue-level flexibility, subcellular feature size and mesh-like networks, into 3D organoids through a 2D-to-3D tissue reconfiguration process during organogenesis. This approach enables longitudinal, tissue-wide, single-cell functional mapping, thereby overcoming the limitations of existing techniques including recording duration, spatial coverage, and the ability to maintain stable contact with the tissue during organoid development. This protocol describes the fabrication and characterization of stretchable mesh nanoelectronics, their electrical performance, their integration with organoids and the acquisition of long-term functional organoid activity requiring multimodal data analysis techniques. Cyborg organoid technology represents a transformative tool for investigating organoid development and function, with potential for improving in vitro disease models, drug screening and personalized medicine. The procedure is suitable for users within a multidisciplinary team with expertise in stem cell biology, tissue engineering, nanoelectronics fabrication, electrophysiology and data science.
Collapse
Affiliation(s)
- Zuwan Lin
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wenbo Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ren Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Qiang Li
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Jaeyong Lee
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Charles Hirschler
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA.
| |
Collapse
|
47
|
Ghirotto B, Gonçalves LE, Ruder V, James C, Gerasimova E, Rizo T, Wend H, Farrell M, Gerez JA, Prymaczok NC, Kuijs M, Shulman M, Hartebrodt A, Prots I, Gessner A, Zunke F, Winkler J, Blumenthal DB, Theis FJ, Riek R, Günther C, Neurath M, Gupta P, Winner B. TNF-α disrupts the malate-aspartate shuttle, driving metabolic rewiring in iPSC-derived enteric neural lineages from Parkinson's Disease patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.644826. [PMID: 40196623 PMCID: PMC11974853 DOI: 10.1101/2025.03.25.644826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Gastrointestinal (GI) dysfunction emerges years before motor symptoms in Parkinson's disease (PD), implicating the enteric nervous system (ENS) in early disease progression. However, the mechanisms linking the PD hallmark protein, α-synuclein (α-syn), to ENS dysfunction - and whether these mechanisms are influenced by inflammation - remains elusive. Using iPSC-derived enteric neural lineages from patients with α-syn triplications, we reveal that TNF-α increases mitochondrial-α-syn interactions, disrupts the malate-aspartate shuttle, and forces a metabolic shift toward glutamine oxidation. These alterations drive mitochondrial dysfunction, characterizing metabolic impairment under cytokine stress. Interestingly, targeting glutamate metabolism with Chicago Sky Blue 6B restores mitochondrial function, reversing TNF-α-driven metabolic disruption. Our findings position the ENS as a central player in PD pathogenesis, establishing a direct link between cytokines, α-syn accumulation, metabolic stress and mitochondrial dysfunction. By uncovering a previously unrecognized metabolic vulnerability in the ENS, we highlight its potential as a therapeutic target for early PD intervention.
Collapse
Affiliation(s)
- Bruno Ghirotto
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- International Max Planck Research School in Physics and Medicine, Erlangen, Germany
| | - Luís Eduardo Gonçalves
- Department of Medicine 1, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Vivien Ruder
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christina James
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Elizaveta Gerasimova
- Dental Clinic 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tania Rizo
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Present address: Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, USA
| | - Holger Wend
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Michaela Farrell
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Juan Atilio Gerez
- Institute of Molecular Physical Sciences, ETH Zürich, Zürich, Switzerland
| | | | - Merel Kuijs
- Institute of Computational Biology, Helmholtz Center, Munich, Germany
- TUM, School of Computation, Information and Technology, Technical University of Munich, Germany
- TUM School of Life Sciences, Technical University of Munich, Germany
| | - Maiia Shulman
- Institute of Computational Biology, Helmholtz Center, Munich, Germany
- TUM, School of Computation, Information and Technology, Technical University of Munich, Germany
- TUM School of Life Sciences, Technical University of Munich, Germany
| | - Anne Hartebrodt
- Biomedical Network Science Lab, Department Artificial Intelligence in Biomedical Engineering , Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Iryna Prots
- Dental Clinic 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arne Gessner
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - David B Blumenthal
- Biomedical Network Science Lab, Department Artificial Intelligence in Biomedical Engineering , Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Center, Munich, Germany
- TUM, School of Computation, Information and Technology, Technical University of Munich, Germany
- TUM School of Life Sciences, Technical University of Munich, Germany
| | - Roland Riek
- Institute of Molecular Physical Sciences, ETH Zürich, Zürich, Switzerland
| | - Claudia Günther
- Department of Medicine 1, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Markus Neurath
- Department of Medicine 1, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Pooja Gupta
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Center of Rare Diseases Erlangen, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
48
|
Menon R, Kimmel PL, Otto EA, Subramanian L, Berthier CC, O' Connor CL, Godfrey B, Naik AS, Sarwal M, Woodle ES, Pyle L, Choi YJ, Ladd P, Sedor JR, Rosas SE, Waikar SS, Bitzer M, Bjornstad P, Hodgin JB, Kretzler M. Not all controls are made equal: Definition of human kidney reference samples by single cell gene expression profiles. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.17.25324134. [PMID: 40166576 PMCID: PMC11957099 DOI: 10.1101/2025.03.17.25324134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Identifying kidney disease mechanisms often requires comparing samples from disease states with healthy reference tissues. However, the effect of variations in sample procurement, storage and donor baseline characteristics of reference samples has thus far not been evaluated. Three distinct kidney reference sample types were evaluated for integrity and injury biomarkers and in their ability to define differentially expressed genes (DEGs) when compared to three different diabetic kidney disease (DKD) states. Unaffected parts of tumor nephrectomies (TN), pre-transplant living donor biopsies (LD), and percutaneous kidney research biopsies from healthy volunteers (HC) served as sources for reference tissue. Single cell gene expression profiles showed differences in the expression of injury or disease markers and the proportion of immune and proximal cell states. TN exhibited the highest expression of early stress response genes. A gene set associated with procurement effect in post-operative biopsies (LD and TN) was identified. An age-associated transcriptional signature was extracted from the reference data. Providing these tools to control for age and tissue procurement effects, immune-related pathways were found to be most enriched in DKD when compared to HC. Energy-related processes were enriched in DEGs from DKD versus LD. TN samples exhibited more underlying pathology than LD. The pathway analyses using the DEGs underscore the importance of accounting for appropriate confounding factors in differential expression analyses between disease and reference samples. Comparable controls are essential for appropriate molecular evaluation of pathologic tissues. TRANSLATIONAL STATEMENT Integrated single-cell data analysis of three reference sample types-needle biopsy from young healthy kidney tissue, pre-perfusion biopsy from transplant kidneys, and cancer-free tissue from tumor-nephrectomies-revealed distinct transcriptional profiles influenced by the biopsy procurement method and age. These differences impacted findings in diabetes-related kidney disease versus reference comparisons highlighting the need and providing the tools to account for these differences in interpreting analyses and identifying disease mechanisms.
Collapse
|
49
|
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
|
50
|
Wang C, Yang M, Zhong Y, Cao K, Wang X, Zhang C, Wang Y, He M, Lu J, Zhang G, Huang Y, Liu H. Immunosuppressive JAG2 + tumor-associated neutrophils hamper PD-1 blockade response in ovarian cancer by mediating the differentiation of effector regulatory T cells. Cancer Commun (Lond) 2025. [PMID: 40120139 DOI: 10.1002/cac2.70021] [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: 06/17/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Tumor-associated neutrophils (TANs) play a critical role in modulating immune responses and exhibit significant heterogeneity. Our previous study demonstrated that jagged canonical Notch ligand 2 (JAG2)+ TANs were associated with an immunosuppressive microenvironment in high-grade serous ovarian cancer (HGSOC), but the underlying mechanism remains unclear. This study aimed to elucidate the role of JAG2+ TANs in tumor immunosuppressive microenvironment in HGSOC. METHODS HGSOC samples were collected, with 274 samples constituting two independent cohorts (training and validation cohorts) and an additional 30 samples utilized to establish patient-derived tumor organoids (PDTOs). We characterized the number and phenotype of JAG2+ TANs by multiplex immunohistochemistry, flow cytometry, and single-cell RNA sequencing (scRNA-seq). We investigated the biological functions of JAG2 in immune evasion using in vitro co-culture systems, flow cytometry, tumor-bearing mouse models, and PDTOs. RESULTS JAG2+ TANs expressed elevated levels of immunosuppressive molecules, including programmed cell death ligand 1 and CD14, and had independent prognostic value for the overall survival of patients with HGSOC. scRNA-seq analysis revealed that JAG2+ TANs exhibited a terminally mature phenotype. The infiltration of JAG2+ TANs was positively correlated with the abundance of effector regulatory T cells (eTregs). Interaction with JAG2+ TANs skewed CD4+ T cells towards an eTreg phenotype, a process that was suppressed by the Notch inhibitor LY3039478 and induced by recombinant Jagged2. Furthermore, we demonstrated that JAG2+ TANs enhanced Notch signaling activation, ultimately promoting recombination signal binding protein for immunoglobulin kappa J region (RBPJ)-induced differentiation of naïve CD4+ T cells into eTregs. Clinically, JAG2+ TANs could serve as a biomarker for assessing immunotherapy resistance in various solid tumors. Pharmacological targeting of Notch signaling with LY3039478 or JAG2 neutralization antibodies enhanced the efficacy of programmed cell death protein 1 (PD-1) monoclonal antibodies (mAbs) in both xenograft and PDTO models. CONCLUSIONS The emergence of JAG2+ TANs is crucial for the differentiation of eTregs, which triggers immune evasion and resistance to anti-PD-1 therapy. Inhibiting Notch signaling with LY3039478 or JAG2 neutralization antibodies may overcome this anti-PD-1 resistance in HGSOC.
Collapse
Affiliation(s)
- Chenyang Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
| | - Moran Yang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
| | - Yujing Zhong
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
| | - Kankan Cao
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
| | - Xueling Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
| | - Chen Zhang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
| | - Yiying Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
| | - Mengdi He
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
| | - Jiaqi Lu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
| | - Guodong Zhang
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
| | - Yan Huang
- Department of Gynecologic Oncology, Shanghai Cancer Center, Fudan University, Shanghai, P. R. China
| | - Haiou Liu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P. R. China
| |
Collapse
|