1
|
Ni G, Sun Y, Jia H, Xiahou Z, Li Y, Zhao F, Zang H. MAZ-mediated tumor progression and immune evasion in hormone receptor-positive breast cancer: Targeting tumor microenvironment and PCLAF+ subtype-specific therapy. Transl Oncol 2025; 52:102280. [PMID: 39805182 DOI: 10.1016/j.tranon.2025.102280] [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: 11/03/2024] [Revised: 12/19/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Breast cancer had been the most frequently diagnosed cancer among women, making up nearly one-third of all female cancers. Hormone receptor-positive breast cancer (HR+BC) was the most prevalent subtype of breast cancer and exhibited significant heterogeneity. Despite advancements in endocrine therapies, patients with advanced HR+BC often faced poor outcomes due to the development of resistance to treatment. Understanding the molecular mechanisms behind this resistance, including tumor heterogeneity and changes in the tumor microenvironment, was crucial for overcoming resistance, identifying new therapeutic targets, and developing more effective personalized treatments. METHODS The study utilized single-cell RNA sequencing (scRNA-seq) data sourced from the Gene Expression Omnibus database and The Cancer Genome Atlas to analyze HR+BC and identify key cellular characteristics. Cell type identification was achieved through Seurat's analytical tools, and subtype differentiation trajectories were inferred using Slingshot. Cellular communication dynamics between tumor cell subtypes and other cells were analyzed with the CellChat. The pySCENIC package was utilized to analyze transcription factors regulatory networks in the identified tumor cell subtypes. The results were verified by in vitro experiments. A risk scoring model was developed to assess patient outcomes. RESULTS This study employed scRNA-seq to conduct a comprehensive analysis of HR+BC tumor subtypes, identifying the C3 PCLAF+ tumor cells subtype, which demonstrated high proliferation and differentiation potential. C3 PCLAF+ tumor cells subtype was found to be closely associated with cancer-associated fibroblasts through the MK signaling pathway, facilitating tumor progression. Additionally, we discovered that MAZ was significantly expressed in C3 PCLAF+ tumor cells subtype, and in vitro experiments confirmed that MAZ knockdown inhibited tumor growth, accentuating its underlying ability as a therapeutic target. Furthermore, we developed a novel prognostic model based on the expression profile of key prognostic genes within the PCLAF+/MAZ regulatory network. This model linked high PCLAF+ tumor risk scores with poor survival outcomes and specific immune microenvironment characteristics. CONCLUSION This study utilized scRNA-seq to reveal the role of the C3 PCLAF+ tumor cells subtype in HR+BC, emphasizing its association with poor prognosis and resistance to endocrine therapies. MAZ, identified as a key regulator, contributed to tumor progression, while the tumor microenvironment had a pivotal identity in immune evasion. The findings underscored the importance of overcoming drug resistance, recognizing novel treatment targets, and crafting tailored diagnosis regimens.
Collapse
Affiliation(s)
- Gaofeng Ni
- Department of Breast Surgery, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai 264003, China
| | - Yuwei Sun
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Hongling Jia
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing 100084, China
| | - Yumeng Li
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Fu Zhao
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Hongyan Zang
- Department of Breast Surgery, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai 264003, China.
| |
Collapse
|
2
|
Ding C, Wang J, Wang J, Niu J, Xiahou Z, Sun Z, Zhao Z, Zeng D. Heterogeneity of cancer-associated fibroblast subpopulations in prostate cancer: Implications for prognosis and immunotherapy. Transl Oncol 2025; 52:102255. [PMID: 39721245 PMCID: PMC11732565 DOI: 10.1016/j.tranon.2024.102255] [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: 10/28/2024] [Revised: 12/03/2024] [Accepted: 12/15/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Prostate cancer stands as the second most common malignancy among men, notorious for its intricate heterogeneity, especially evident in metastatic disease. This complexity presents substantial challenges in treatment efficacy and patient prognosis. OBJECTIVE This study endeavors to elucidate the multifaceted roles of cancer-associated fibroblasts within the tumor microenvironment of prostate cancer, with a focus on their implications for disease prognosis and the potential for novel immunotherapeutic strategies. METHODS Leveraging advanced single-cell RNA sequencing technology, we meticulously characterized the diverse CAF subpopulations within prostate cancer samples. Our analysis identified four predominant subsets: C0 IER2+, C1 ABCA8+, C2 ABI3BP+, and C3 MEOX2+. We conducted comprehensive gene expression profiling to construct a robust prognostic model reflecting the clinical relevance of these subpopulations. RESULTS C1 ABCA8+ fibroblasts demonstrated heightened proliferative activity, underscoring their pivotal role in fostering tumor growth and metastasis via intricate signaling pathways. In vitro experiments verified that the T transcription factor NFAT5 of C1 ABCA8+ fibroblasts subpopulation was knocked down in LNCaP clone FGC and 22Rv1 cell lines, which was closely related to the proliferation of PC. Moreover, we identified key genes linked to patient outcomes and immune landscape alterations, reinforcing the prognostic significance of CAF characteristics in this context. CONCLUSION This investigation illuminates the critical potential of targeting CAFs to augment immunotherapeutic approaches in prostate cancer. Our findings contribute to a deeper understanding of the TME's complexity, advocating for further exploration into CAF-targeted therapies aimed at enhancing treatment responses and ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Chen Ding
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, 136 Jingzhou Street, Xiangyang, Hubei 441021, PR China
| | - Jiange Wang
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, 136 Jingzhou Street, Xiangyang, Hubei 441021, PR China
| | - Jie Wang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, China; Department of Urology, The Second People's Hospital of Meishan City, Meishan, Sichuan, China
| | - Jiqiang Niu
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Zhou Sun
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130000, China.
| | - Zhenzhen Zhao
- The first clinical medical college of Shandong university of Traditional Chinese Medicine, Jinan 250014, China.
| | - Dongyang Zeng
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, 136 Jingzhou Street, Xiangyang, Hubei 441021, PR China.
| |
Collapse
|
3
|
Leshem R, Sefton KN, Wong CW, Lin IH, Isaac DT, Niepel M, Hurlstone A. Combined PARP14 inhibition and PD-1 blockade promotes cytotoxic T cell quiescence and modulates macrophage polarization in relapsed melanoma. J Immunother Cancer 2025; 13:e010683. [PMID: 39870492 DOI: 10.1136/jitc-2024-010683] [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] [Accepted: 01/09/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Programmed cell death 1 (PD-1) signaling blockade by immune checkpoint inhibitors (ICI) effectively restores immune surveillance to treat melanoma. However, chronic interferon-gamma (IFNγ)-induced immune homeostatic responses in melanoma cells contribute to immune evasion and acquired resistance to ICI. Poly ADP ribosyl polymerase 14 (PARP14), an IFNγ-responsive gene product, partially mediates IFNγ-driven resistance. PARP14 inhibition prolongs PD-1 blockade responses in preclinical models, but fails to achieve full tumor clearance, suggesting the involvement of additional resistance mechanisms. METHODS We identified a robust PARP14 catalytic inhibitor gene signature and evaluated its association with patient survival. Using preclinical models and single-cell RNA sequencing, we investigated immune and tumor cell adaptations to PARP14 inhibition combined with PD-1 blockade. RESULTS Combining PARP14 inhibition and PD-1 blockade suppressed tumor-associated macrophages while increasing proinflammatory memory macrophages. Moreover, this combination mitigated the terminal exhaustion of cytotoxic T cells by inducing a quiescent state, thereby preserving functionality. Despite the enhanced immune responses, tumor cells developed adaptive resistance by engaging alternative immune evasion pathways. CONCLUSIONS Although adaptive resistance mechanisms re-emerge, PARP14 inhibition combined with PD-1 blockade offers a promising strategy to enhance treatment outcomes and overcome ICI resistance in melanoma, as immune cells are primed for further therapeutic interventions that leverage the quiescent state.
Collapse
Affiliation(s)
- Rotem Leshem
- Division of Infection, Immunity and Respiratory Medicine, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
- School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Kieran Neil Sefton
- Division of Infection, Immunity and Respiratory Medicine, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
- School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Chun Wai Wong
- Division of Infection, Immunity and Respiratory Medicine, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
- School of Biological Sciences, The University of Manchester, Manchester, UK
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - I-Hsuan Lin
- Bioinformatics Core Facility, The University of Manchester, Manchester, UK
| | | | - Mario Niepel
- Ribon Therapeutics, Cambridge, Massachusetts, USA
| | - Adam Hurlstone
- Division of Infection, Immunity and Respiratory Medicine, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
- School of Biological Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
4
|
Sessions GA, Loops MV, Diekman BO, Purvis JE. Multiplexed single-cell imaging reveals diverging subpopulations with distinct senescence phenotypes during long-term senescence induction. GeroScience 2025:10.1007/s11357-024-01503-7. [PMID: 39849264 DOI: 10.1007/s11357-024-01503-7] [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: 10/30/2024] [Accepted: 12/26/2024] [Indexed: 01/25/2025] Open
Abstract
Cellular senescence is a phenotypic state that contributes to the progression of age-related disease through secretion of pro-inflammatory factors known as the senescence-associated secretory phenotype (SASP). Understanding the process by which healthy cells become senescent and develop SASP factors is critical for improving the identification of senescent cells and, ultimately, understanding tissue dysfunction. Here, we reveal how the duration of cellular stress modulates the SASP in distinct subpopulations of senescent cells. We used multiplex, single-cell imaging to build a proteomic map of senescence induction in human epithelial cells induced to senescence over the course of 31 days. We map how the expression of SASP proteins increases alongside other known senescence markers such as p53, p21, and p16INK4a. The aggregated population of cells responded to etoposide with an accumulation of stress response factors over the first 11 days, followed by a plateau in most proteins. At the single-cell level, however, we identified two distinct senescence cell populations, one defined primarily by larger nuclear area and the second by higher protein concentrations. Trajectory inference suggested that cells took one of two discrete molecular paths from unperturbed healthy cells, through a common transitional subpopulation, and ending at the discrete terminal senescence phenotypes. Our results underscore the importance of using single-cell proteomics to identify the mechanistic pathways governing the transition from senescence induction to a mature state of senescence characterized by the SASP.
Collapse
Affiliation(s)
- Garrett A Sessions
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Madeline V Loops
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL, 60637, USA
| | - Brian O Diekman
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, 4111-A Thurston, Campus Box #7280, Chapel Hill, NC, 27599, USA.
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, 27695, USA.
| | - Jeremy E Purvis
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Genetics, The University of North Carolina School of Medicine, 11018C Mary Ellen Jones, Campus Box #7264, Chapel Hill, NC, 27599, USA.
- Computational Medicine Program, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
5
|
Onder L, Papadopoulou C, Lütge A, Cheng HW, Lütge M, Perez-Shibayama C, Gil-Cruz C, De Martin A, Kurz L, Cadosch N, Pikor NB, Rodriguez R, Born D, Jochum W, Leskow P, Dutly A, Robinson MD, Ludewig B. Fibroblastic reticular cells generate protective intratumoral T cell environments in lung cancer. Cell 2025; 188:430-446.e20. [PMID: 39566495 DOI: 10.1016/j.cell.2024.10.042] [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: 03/18/2024] [Revised: 08/28/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024]
Abstract
Stringent control of T cell activity in the tumor microenvironment is essential for the generation of protective antitumor immunity. However, the identity, differentiation, and functions of the cells that create critical fibroblastic niches promoting tumor-infiltrating T cells remain elusive. Here, we show that CCL19-expressing fibroblastic reticular cells (FRCs) generate interconnected T cell environments (TEs) in human non-small cell lung cancer, including tertiary lymphoid structures and T cell tracks. Analysis of the FRC-T cell interactome in TEs indicated molecular networks regulating niche-specific differentiation of CCL19-expressing fibroblasts and T cell activation pathways. Single-cell transcriptomics and cell fate-mapping analyses in mice confirmed that FRCs in TEs originate from mural and adventitial progenitors. Ablation of intratumoral FRC precursors decreased antitumor T cell activity, resulting in reduced tumor control during coronavirus vector-based immunotherapy. In summary, specialized FRC niches in the tumor microenvironment govern the quality and extent of antitumor T cell immunity.
Collapse
Affiliation(s)
- Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland.
| | - Chrysa Papadopoulou
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Almut Lütge
- Department of Molecular Life Sciences and SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich 8057, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | | | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Angelina De Martin
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Lisa Kurz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Nadine Cadosch
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Natalia B Pikor
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland; Institute of Microbiology and Immunology, ETH Zurich, Zurich 8093, Switzerland
| | - Regulo Rodriguez
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Diana Born
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Wolfram Jochum
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Pawel Leskow
- Department of Thoracic Surgery, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Andre Dutly
- Department of Thoracic Surgery, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Mark D Robinson
- Department of Molecular Life Sciences and SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich 8057, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland; University Heart Center, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland; Center for Translational and Experimental Cardiology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland.
| |
Collapse
|
6
|
Volinia S, Terrazzan A, Kaminski TS, Jadzewski K, Reali E, Bianchi N, Palatini J. Circulating tumor cells share RNA modules with early embryo trophectoderm and with metastatic cancer. Cancer Commun (Lond) 2025. [PMID: 39853715 DOI: 10.1002/cac2.12664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Affiliation(s)
- Stefano Volinia
- Department of Translational Medicine, Laboratory for Advanced Therapy Technologies, University of Ferrara, Ferrara, Italy
- Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
- Centre of New Technology, University of Warsaw, Warsaw, Poland
| | - Anna Terrazzan
- Department of Translational Medicine, Laboratory for Advanced Therapy Technologies, University of Ferrara, Ferrara, Italy
| | - Tomasz S Kaminski
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Krystian Jadzewski
- Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Eva Reali
- Department of Translational Medicine, Laboratory for Advanced Therapy Technologies, University of Ferrara, Ferrara, Italy
| | - Nicoletta Bianchi
- Department of Translational Medicine, Laboratory for Advanced Therapy Technologies, University of Ferrara, Ferrara, Italy
| | - Jeff Palatini
- Centre of New Technology, University of Warsaw, Warsaw, Poland
| |
Collapse
|
7
|
Shin D, Gong JR, Jeong SD, Cho Y, Kim HP, Kim TY, Cho KH. Attractor Landscape Analysis Reveals a Reversion Switch in the Transition of Colorectal Tumorigenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412503. [PMID: 39840939 DOI: 10.1002/advs.202412503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/27/2024] [Indexed: 01/23/2025]
Abstract
A cell fate change such as tumorigenesis incurs critical transition. It remains a longstanding challenge whether the underlying mechanism can be unraveled and a molecular switch that can reverse such transition is found. Here a systems framework, REVERT, is presented with which can reconstruct the core molecular regulatory network model and a reversion switch based on single-cell transcriptome data over the transition process is identified. The usefulness of REVERT is demonstrated by applying it to single-cell transcriptome of patient-derived matched organoids of colon cancer and normal colon. REVERT is a generic framework that can be applied to investigate various cell fate transition phenomena.
Collapse
Affiliation(s)
- Dongkwan Shin
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- Research Institute, National Cancer Center, Goyang, 10408, Republic of Korea
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, 10408, Republic of Korea
| | - Jeong-Ryeol Gong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seoyoon D Jeong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Youngwon Cho
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hwang-Phill Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 03080, Republic of Korea
| | - Tae-You Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 03080, Republic of Korea
| | - Kwang-Hyun Cho
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| |
Collapse
|
8
|
Nouri N. Single-cell RNA-seq data augmentation using generative Fourier transformer. Commun Biol 2025; 8:113. [PMID: 39843603 PMCID: PMC11754799 DOI: 10.1038/s42003-025-07552-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) provides a powerful tool for dissecting cellular complexity and heterogeneity. However, its full potential to achieve statistically reliable conclusions is often constrained by the limited number of cells profiled, particularly in studies of rare diseases, specialized tissues, and uncommon cell types. Deep learning-based generative models (GMs) designed to address data scarcity often face similar limitations due to their reliance on pre-training or fine-tuning, inadvertently perpetuating a cycle of data inadequacy. To overcome this obstacle, we introduce scGFT (single-cell Generative Fourier Transformer), a train-free, cell-centric GM adept at synthesizing single cells that exhibit natural gene expression profiles present within authentic datasets. Using both simulated and experimental data, we demonstrate the mathematical rigor of scGFT and validate its ability to synthesize cells that preserve the intrinsic characteristics delineated in scRNA-seq data. Moreover, comparisons of scGFT with leading neural network-based GMs highlight its superior performance, driven by its analytical mechanism. By streamlining single-cell data augmentation, scGFT offers a scalable solution to mitigate data scarcity in cell-targeted research.
Collapse
Affiliation(s)
- Nima Nouri
- Precision Medicine and Computational Biology, Sanofi, Cambridge, MA, 02141, USA.
| |
Collapse
|
9
|
Chen X, Zhang C, Peng F, Wu L, Zhuo D, Wang L, Zhang M, Li Z, Tian L, Jie Y, Huang Y, Yang X, Li X, Lei F, Cheng Y. Identification of glutamine as a potential therapeutic target in dry eye disease. Signal Transduct Target Ther 2025; 10:27. [PMID: 39837870 PMCID: PMC11751114 DOI: 10.1038/s41392-024-02119-1] [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: 08/19/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/30/2025] Open
Abstract
Dry eye disease (DED) is a prevalent inflammatory condition significantly impacting quality of life, yet lacks effective pharmacological therapies. Herein, we proposed a novel approach to modulate the inflammation through metabolic remodeling, thus promoting dry eye recovery. Our study demonstrated that co-treatment with mesenchymal stem cells (MSCs) and thymosin beta-4 (Tβ4) yielded the best therapeutic outcome against dry eye, surpassing monotherapy outcomes. In situ metabolomics through matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) revealed increased glutamine levels in cornea following MSC + Tβ4 combined therapy. Inhibition of glutamine reversed the anti-inflammatory, anti-apoptotic, and homeostasis-preserving effects observed with combined therapy, highlighting the critical role of glutamine in dry eye therapy. Clinical cases and rodent model showed elevated expression of glutaminase (GLS1), an upstream enzyme in glutamine metabolism, following dry eye injury. Mechanistic studies indicated that overexpression and inhibition of GLS1 counteracted and enhanced, respectively, the anti-inflammatory effects of combined therapy, underscoring GLS1's pivotal role in regulating glutamine metabolism. Furthermore, single-cell sequencing revealed a distinct subset of pro-inflammatory and pro-fibrotic corneal epithelial cells in the dry eye model, while glutamine treatment downregulated those subclusters, thereby reducing their inflammatory cytokine secretion. In summary, glutamine effectively ameliorated inflammation and the occurrence of apoptosis by downregulating the pro-inflammatory and pro-fibrotic corneal epithelial cells subclusters and the related IκBα/NF-κB signaling. The present study suggests that glutamine metabolism plays a critical, previously unrecognized role in DED and proposes an attractive strategy to enhance glutamine metabolism by inhibiting the enzyme GLS1 and thus alleviating inflammation-driven DED progression.
Collapse
Affiliation(s)
- Xiaoniao Chen
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China.
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China.
| | - Chuyue Zhang
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China
| | - Fei Peng
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China
| | - Lingling Wu
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China
| | - Deyi Zhuo
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Liqiang Wang
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China
| | - Min Zhang
- Department of Nephrology, the First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, China
| | - Zhaohui Li
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lei Tian
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ying Jie
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yifei Huang
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xinji Yang
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaoqi Li
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Fengyang Lei
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yu Cheng
- Department of Ophthalmology, the Third Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
10
|
Chen Y, Hou Y, Zeng Q, Wang I, Shang M, Shin K, Hemauer C, Xing X, Kang J, Zhao G, Wang T. Common and specific gene regulatory programs in zebrafish caudal fin regeneration at single-cell resolution. Genome Res 2025; 35:202-218. [PMID: 39809530 DOI: 10.1101/gr.279372.124] [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: 03/18/2024] [Accepted: 11/04/2024] [Indexed: 01/16/2025]
Abstract
Following amputation, zebrafish regenerate their injured caudal fin through lineage-restricted reprogramming. Although previous studies have charted various genetic and epigenetic dimensions of this process, the intricate gene regulatory programs shared by, or unique to, different regenerating cell types remain underinvestigated. Here, we mapped the regulatory landscape of fin regeneration by applying paired snRNA-seq and snATAC-seq on uninjured and regenerating fins. This map delineates the regulatory dynamics of predominant cell populations at multiple stages of regeneration. We observe a marked increase in the accessibility of chromatin regions associated with regenerative and developmental processes at 1 dpa, followed by a gradual closure across major cell types at later stages. This pattern is distinct from that of transcriptomic dynamics, which is characterized by several waves of gene upregulation and downregulation. We identified and in vivo validated cell-type-specific and position-specific regeneration-responsive enhancers and constructed regulatory networks by cell type and stage. Our single-cell resolution transcriptomic and chromatin accessibility map across regenerative stages provides new insights into regeneration regulatory mechanisms and serves as a valuable resource for the community.
Collapse
Affiliation(s)
- Yujie Chen
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yiran Hou
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Qinglin Zeng
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Irene Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Meiru Shang
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Kwangdeok Shin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin 53705, USA
| | - Christopher Hemauer
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin 53705, USA
| | - Guoyan Zhao
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
11
|
Zhang Y, Lu Z, Guo J, Wang Q, Zhang X, Yang H, Li X. Advanced Carriers for Precise Delivery and Therapeutic Mechanisms of Traditional Chinese Medicines: Integrating Spatial Multi-Omics and Delivery Visualization. Adv Healthc Mater 2025:e2403698. [PMID: 39828637 DOI: 10.1002/adhm.202403698] [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: 10/17/2024] [Revised: 12/01/2024] [Indexed: 01/22/2025]
Abstract
The complex composition of traditional Chinese medicines (TCMs) has posed challenges for in-depth study and global application, despite their abundance of bioactive compounds that make them valuable resources for disease treatment. To overcome these obstacles, it is essential to modernize TCMs by focusing on precise disease treatment. This involves elucidating the structure-activity relationships within their complex compositions, ensuring accurate in vivo delivery, and monitoring the delivery process. This review discusses the research progress of TCMs in precision disease treatment from three perspectives: spatial multi-omics technology for precision therapeutic activity, carrier systems for precise in vivo delivery, and medical imaging technology for visualizing the delivery process. The aim is to establish a novel research paradigm that advances the precision therapy of TCMs.
Collapse
Affiliation(s)
- Yusheng Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
| | - Zhiguo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Jing Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Qing Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, P. R. China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Hongjun Yang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, China Academy of Chinese Medical Sciences, Beijing, 100029, P. R. China
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
| |
Collapse
|
12
|
Marchand V, Laplane L, Valensi L, Plo I, Aglave M, Silvin A, Pasquier F, Porteu F, Vainchenker W, Selimoglu-Buet D, Droin N, Raslova H, Marcel V, Diaz JJ, Fontenay M, Solary E. Monocytes generated by interleukin-6-treated human hematopoietic stem and progenitor cells secrete calprotectin that inhibits erythropoiesis. iScience 2025; 28:111522. [PMID: 39811665 PMCID: PMC11732210 DOI: 10.1016/j.isci.2024.111522] [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: 06/20/2024] [Revised: 10/02/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Elevated circulating levels of calprotectin (CAL), the S100A8/A9 heterodimer, are biomarkers of severe systemic inflammation. Here, we investigate the effects of CAL on early human hematopoiesis. CAL demonstrates limited impact on gene expression in stem and progenitor cells, in contrast with interleukin-6 (IL6), which promotes the expression of the S100A8 and S100A9 genes in hematopoietic progenitors and the generation of monocytes that release CAL. The main target of CAL is an erythroid-megakaryocyte progenitor (EMP) subset. CAL prevents both erythropoietin-driven differentiation of healthy progenitors and JAK2-V617F-driven erythropoiesis. In the context of JAK2-V617F, CAL also promotes the expression of S100A8 and S100A9 genes in monocytes. The signature of CAL effects is detected in the bone marrow progenitors of patients with myeloid malignancy or severe infection. These results position CAL as a mediator of IL6 effects on triggering anemia during inflammation, an effect that is amplified in the context of JAK2-V617F-driven hematopoiesis.
Collapse
Affiliation(s)
- Valentine Marchand
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Lucie Laplane
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
- CNRS 8590, Université Paris 1 Panthéon-Sorbonne, Paris, France
| | - Louis Valensi
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Isabelle Plo
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Marine Aglave
- AMMICa, INSERM US 23, CNRS UMS 3655, Gustave Roussy Cancer Center, Villejuif, France
| | - Aymeric Silvin
- INSERM U1108, Gustave Roussy Cancer Center, Villejuif, France
| | - Florence Pasquier
- Department of Hematology, Gustave Roussy Cancer Center, Villejuif, France
| | - Françoise Porteu
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - William Vainchenker
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | | | - Nathalie Droin
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
- AMMICa, INSERM US 23, CNRS UMS 3655, Gustave Roussy Cancer Center, Villejuif, France
| | - Hana Raslova
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Virginie Marcel
- Inserm U1052, CNRS UMR5286 Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Jean-Jacques Diaz
- Inserm U1052, CNRS UMR5286 Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Michaela Fontenay
- Université Paris Cité, Institut Cochin, CNRS UMR 8104, INSERM U1016, Paris, France
- Laboratory of Excellence for Red Blood Cells, GR-Ex, Paris, France
| | - Eric Solary
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| |
Collapse
|
13
|
Lo Tartaro D, Aramini B, Masciale V, Paschalidis N, Lofaro FD, Neroni A, Borella R, Santacroce E, Ciobanu AL, Samarelli AV, Boraldi F, Quaglino D, Dubini A, Gaudio M, Manzotti G, Reggiani F, Torricelli F, Ciarrocchi A, Neri A, Bertolini F, Dominici M, Filosso PL, Stella F, Gibellini L, De Biasi S, Cossarizza A. Metabolically activated and highly polyfunctional intratumoral VISTA + regulatory B cells are associated with tumor recurrence in early-stage NSCLC. Mol Cancer 2025; 24:16. [PMID: 39810191 PMCID: PMC11730485 DOI: 10.1186/s12943-024-02209-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
B cells have emerged as central players in the tumor microenvironment (TME) of non-small cell lung cancer (NSCLC). However, although there is clear evidence for their involvement in cancer immunity, scanty data exist on the characterization of B cell phenotypes, bioenergetic profiles and possible interactions with T cells in the context of NSCLC. In this study, using polychromatic flow cytometry, mass cytometry, and spatial transcriptomics we explored the intricate landscape of B cell phenotypes, bioenergetics, and their interaction with T cells in NSCLC. Our analysis revealed that TME contains diverse B cell clusters, including VISTA+ Bregs, with distinct metabolic and functional profiles. Target liquid chromatography-tandem mass spectrometry confirmed the expression of VISTA on B cells. VISTA+ Bregs displayed high metabolic demand and were able to produce different cytokines, including interleukin (IL)-10, transforming growth factor (TGF)-β, IL-6, tumor necrosis factor (TNF), and granulocyte-macrophage colony-stimulating factor (GM-CSF). Spatial analysis showed colocalization of B cells with CD4+/CD8+ T lymphocytes in TME. The computational analysis of intercellular communications that links ligands to target genes, performed by NicheNet, predicted B-T interactions via VISTA-PSGL-1 axis. Colocalization analyses revealed that PSGL-1 T cells and VISTA+ B cells are adjacent in the TME. Notably, tumor infiltrating CD8+ T cells expressing PSGL-1 exhibited enhanced metabolism and cytotoxicity. In NSCLC patients, prediction analysis performed by PENCIL revealed the presence of an association between PSGL-1+CD8+ T cells and VISTA+ Bregs with lung recurrence. Our findings suggest a potential interaction between Bregs and T cells through the VISTA-PSGL-1 axis, that could influence NSCLC recurrence.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/immunology
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/immunology
- Lung Neoplasms/genetics
- Tumor Microenvironment/immunology
- B7 Antigens/metabolism
- B7 Antigens/genetics
- B-Lymphocytes, Regulatory/metabolism
- B-Lymphocytes, Regulatory/immunology
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/metabolism
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Female
- Male
- Neoplasm Staging
- Middle Aged
- Cytokines/metabolism
- Aged
- Biomarkers, Tumor/metabolism
Collapse
Affiliation(s)
- Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences - DIMEC, University of Bologna, G.B. Morgagni -L. Pierantoni Hospital, Forlì, Italy
| | - Valentina Masciale
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy
| | | | | | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy
| | - Elena Santacroce
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy
| | - Alin Liviu Ciobanu
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy
| | - Anna Valeria Samarelli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy
| | - Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Dubini
- Division of Pathology, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Michele Gaudio
- Division of Pathology, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Gloria Manzotti
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Francesca Reggiani
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Federica Torricelli
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Antonino Neri
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Federica Bertolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy
- Division of Oncology and Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy
- Division of Oncology and Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Pier Luigi Filosso
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Franco Stella
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences - DIMEC, University of Bologna, G.B. Morgagni -L. Pierantoni Hospital, Forlì, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy.
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, via Campi, 287, Modena, 41125, Italy.
- National Institute for Cardiovascular Research, Bologna, Italy.
| |
Collapse
|
14
|
Niethamer TK, Planer JD, Morley MP, Babu A, Zhao G, Basil MC, Cantu E, Frank DB, Diamond JM, Nottingham AN, Li S, Sharma A, Hallquist H, Levin LI, Zhou S, Vaughan AE, Morrisey EE. Longitudinal single-cell profiles of lung regeneration after viral infection reveal persistent injury-associated cell states. Cell Stem Cell 2025:S1934-5909(24)00441-7. [PMID: 39818203 DOI: 10.1016/j.stem.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/12/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
Functional regeneration of the lung's gas exchange surface following injury requires the coordination of a complex series of cell behaviors within the alveolar niche. Using single-cell transcriptomics combined with lineage tracing of proliferating progenitors, we examined mouse lung regeneration after influenza injury, demonstrating an asynchronously phased response across different cellular compartments. This longitudinal atlas of injury responses has produced a catalog of transient and persistent transcriptional alterations in cells as they transit across axes of differentiation. These cell states include an injury-induced capillary endothelial cell (iCAP) that arises after injury, persists indefinitely, and shares hallmarks with developing lung endothelium and endothelial aberrations found in degenerative human lung diseases. This dataset provides a foundational resource to understand the complexity of cellular and molecular responses to injury and correlations to responses found in human development and disease.
Collapse
Affiliation(s)
- Terren K Niethamer
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| | - Joseph D Planer
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael P Morley
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Apoorva Babu
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gan Zhao
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria C Basil
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward Cantu
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David B Frank
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Pediatric Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joshua M Diamond
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ana N Nottingham
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shanru Li
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arnav Sharma
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Hannah Hallquist
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lillian I Levin
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Su Zhou
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew E Vaughan
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
Yang S, Yu B, Zhang Q, Zhang Y, Fu L, Zhou B, Wu H, Li J, Gong S. Amantadine modulates novel macrophage phenotypes to enhance neural repair following spinal cord injury. J Transl Med 2025; 23:60. [PMID: 39806436 PMCID: PMC11726942 DOI: 10.1186/s12967-024-05916-y] [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: 07/16/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Spinal cord injury (SCI) triggers a complex inflammatory response that impedes neural repair and functional recovery. The modulation of macrophage phenotypes is thus considered a promising therapeutic strategy to mitigate inflammation and promote regeneration. METHODS We employed microarray and single-cell RNA sequencing (scRNA-seq) to investigate gene expression changes and immune cell dynamics in mice following crush injury at 3 and 7 days post-injury (dpi). High-dimensional gene co-expression network analysis (hdWGCNA) and slingshot trajectory analysis were employed to identify key gene modules and macrophage differentiation pathways. Subsequently, immunofluorescence staining, flow cytometry, and western blotting were performed to validate the identified effects of amantadine on macrophage differentiation and inflammation. RESULTS To elucidate the molecular mechanisms underlying the injury response at the transcriptional level, we performed a microarray analysis followed by gene set enrichment analysis (GSEA). The results revealed that pathways related to phagocytosis and macrophage activation are significantly involved post-injury, shedding light on the regulatory role of macrophages in SCI repair. To further investigate macrophage dynamics within the injured spinal cord, we conducted scRNA-Seq, identifying three distinct macrophage subtypes: border-associated macrophages (BAMs), inflammatory macrophages (IMs), and chemotaxis-inducing macrophages (CIMs). Trajectory analysis suggested a differentiation pathway from Il-1b+ IMs to Mrc1+ BAMs, and subsequently to Arg1+ CIMs, indicating a potential maturation process. Given the importance of these pathways in the injury response, we utilized molecular docking to hypothesize that amantadine might modulate this process. Subsequent in vitro and in vivo experiments demonstrated that amantadine reduces Il-1b+ IMs and facilitates the transition to Mrc1+ BAMs and Arg1+ CIMs, likely through modulation of the HIF-1α and NF-κB pathways. This modulation promotes neural regeneration and enhances functional recovery following SCI. CONCLUSIONS Amantadine modulates macrophage phenotypes following SCI, reduces early inflammatory responses, and enhances neural function recovery. These findings highlight the therapeutic potential of amantadine as a treatment for SCI, and provide a foundation for future translational research into its clinical applications.
Collapse
Affiliation(s)
- Shijie Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Beibei Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Qing Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yongfeng Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Longhui Fu
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Bisheng Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Haining Wu
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jianzhong Li
- Department of Thoracic Surgery, Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China.
| | - Shouping Gong
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China.
- Xi'an Medical University, Xi'an, China.
| |
Collapse
|
16
|
Li X, Hegarty K, Lin F, Chang JL, Abdalla A, Dhanabalan K, Solomevich SO, Song W, Roder K, Yao C, Lu W, Carmeliet P, Choudhary G, Dennery PA, Yao H. Endothelial Cpt1a Inhibits Neonatal Hyperoxia-Induced Pulmonary Vascular Remodeling by Repressing Endothelial-Mesenchymal Transition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415824. [PMID: 39799584 DOI: 10.1002/advs.202415824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/15/2024] [Indexed: 01/15/2025]
Abstract
Pulmonary hypertension (PH) increases the mortality of preterm infants with bronchopulmonary dysplasia (BPD). There are no curative therapies for this disease. Lung endothelial carnitine palmitoyltransferase 1a (Cpt1a), the rate-limiting enzyme of the carnitine shuttle system, is reduced in a rodent model of BPD. It is unknown whether endothelial Cpt1a reduction causes pulmonary vascular (PV) remodeling. The latter can be the result of endothelial-mesenchymal transition (EndoMT). Here, endothelial cell (EC)-specific Cpt1a KO and WT mice (<12 h old) are exposed to hyperoxia (70% O2) for 14 days and allow them to recover in normoxia until postnatal day 28. Hyperoxia causes PH, which is aggravated in EC-specific Cpt1a KO mice. Upregulating endothelial Cpt1a expression inhibits hyperoxia-induced PV remodeling. Hyperoxia causes lung EndoMT, detected by immunofluorescence, scRNA-sequencing, and EC lineage tracing, which is further increased in EC-specific Cpt1a KO mice. Blocking EndoMT inhibits hyperoxia-induced PV remodeling. Male mice under the same high oxygen conditions develop a higher degree of PH than females, which is associated with reduced endothelial Cpt1a expression. Conclusively, neonatal hyperoxia causes PH by decreasing endothelial Cpt1a expression and upregulating EndoMT. This provides a valuable strategy for developing targeted therapies by upregulating endothelial Cpt1a levels or inhibiting EndoMT to treat BPD-associated PH.
Collapse
Affiliation(s)
- Xiaoyun Li
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI, 02912, USA
- Providence VA Medical Center, Providence, RI, 02908, USA
- Division of Cardiology, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Katy Hegarty
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI, 02912, USA
| | - Fanjie Lin
- State Key Laboratory of Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Jason L Chang
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI, 02912, USA
| | - Amro Abdalla
- Providence VA Medical Center, Providence, RI, 02908, USA
| | - Karthik Dhanabalan
- Division of Cardiology, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Sergey O Solomevich
- Division of Cardiology, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Wenliang Song
- Division of Cardiology, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Karim Roder
- Division of Cardiology, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Chenrui Yao
- College of Arts & Sciences, Boston University, Boston, MA, 02215, USA
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute, KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Brussels, 3000, Belgium
- Center for Biotechnology, Khalifa University, Abu Dhabi, 127788, UAE
| | - Gaurav Choudhary
- Providence VA Medical Center, Providence, RI, 02908, USA
- Division of Cardiology, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Phyllis A Dennery
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI, 02912, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Hongwei Yao
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI, 02912, USA
- Providence VA Medical Center, Providence, RI, 02908, USA
- Division of Cardiology, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| |
Collapse
|
17
|
Maulding ND, Zou J, Zhou W, Metcalfe C, Stuart JM, Ye X, Hafner M. Transformer-based modeling of Clonal Selection and Expression Dynamics reveals resistance mechanisms in breast cancer. NPJ Syst Biol Appl 2025; 11:5. [PMID: 39794360 PMCID: PMC11723929 DOI: 10.1038/s41540-024-00485-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Understanding transcriptional heterogeneity in cancer cells and its implication for treatment response is critical to identify how resistance occurs and may be targeted. Such heterogeneity can be captured by in vitro studies through clonal barcoding methods. We present TraCSED (Transformer-based modeling of Clonal Selection and Expression Dynamics), a dynamic deep learning approach for modeling clonal selection. Using single-cell gene expression and the fitness of barcoded clones, TraCSED identifies interpretable gene programs and the time points at which they are associated with clonal selection. When applied to cells treated with either giredestrant, a selective estrogen receptor (ER) antagonist and degrader, or palbociclib, a CDK4/6 inhibitor, pathways dynamically associated with resistance are revealed. For example, ER activity is associated with positive selection around day four under palbociclib treatment and this adaptive response can be suppressed by combining the drugs. Yet, in the combination treatment, one clone still emerged. Clustering based on partial least squares regression found that high baseline expression of both SNHG25 and SNCG genes was the primary marker of positive selection to co-treatment and thus potentially associated with innate resistance - an aspect that traditional differential analysis methods missed. In conclusion, TraCSED enables associating features with phenotypes in a time-dependent manner from scRNA-seq data.
Collapse
Affiliation(s)
- Nathan D Maulding
- gRED Computational Sciences, Genentech Inc, South San Francisco, CA, USA
- Department of Biomolecular Engineering and Bioinformatics, UC Santa Cruz, Santa Cruz, CA, USA
| | - Jun Zou
- Discovery Oncology, Genentech Inc, South San Francisco, CA, USA
| | - Wei Zhou
- Discovery Oncology, Genentech Inc, South San Francisco, CA, USA
| | - Ciara Metcalfe
- Discovery Oncology, Genentech Inc, South San Francisco, CA, USA
| | - Joshua M Stuart
- Department of Biomolecular Engineering and Bioinformatics, UC Santa Cruz, Santa Cruz, CA, USA
| | - Xin Ye
- Discovery Oncology, Genentech Inc, South San Francisco, CA, USA
| | - Marc Hafner
- gRED Computational Sciences, Genentech Inc, South San Francisco, CA, USA.
- Discovery Oncology, Genentech Inc, South San Francisco, CA, USA.
| |
Collapse
|
18
|
Singh A, Ratnapriya R. Integration of multiomic data identifies core-module of inherited-retinal diseases. Hum Mol Genet 2025:ddaf001. [PMID: 39797395 DOI: 10.1093/hmg/ddaf001] [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: 10/09/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
Human diseases with similar phenotypes can be interconnected through shared biological pathways, genes, or molecular mechanisms. Inherited retinal diseases (IRDs) cause photoreceptor dysfunction due to mutations in approximately 300 genes, affecting visual transduction, photoreceptor morphogenesis, and transcription factors, suggesting common pathobiological mechanisms. This study examined the functional relationship between known IRDs genes by integrating binding sites and gene expression data from the key photoreceptor transcription factors (TFs), Crx and Nrl. We show that the targets of these TFs were enriched in IRDs causal genes. Co-expression network analysis revealed that IRD-centric networks were disrupted when Crx and Nrl were knocked out. Finally, we identified a highly connected core module comprising 14 IRD and 39 target genes, of which 29 were dysregulated in the rod photoreceptors of the four IRD mouse models. These findings offer a network-based interpretation of IRDs, aiding in the identification of common mechanisms, prioritizing genes for novel disease gene identification, and informing the development of gene-agnostic therapies for IRDs.
Collapse
Affiliation(s)
- Ajeet Singh
- Department of Ophthalmology, Baylor College of Medicine, 6565 Fannin St, NC205, Houston, TX 77030 United States
| | - Rinki Ratnapriya
- Department of Ophthalmology, Baylor College of Medicine, 6565 Fannin St, NC205, Houston, TX 77030 United States
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, 6565 Fannin St, NC205, Houston, TX 77030 United States
| |
Collapse
|
19
|
Peng B, Wang Q, Zhang F, Shen H, Du P. Mouse totipotent blastomere-like cells model embryogenesis from zygotic genome activation to post implantation. Cell Stem Cell 2025:S1934-5909(24)00446-6. [PMID: 39826539 DOI: 10.1016/j.stem.2024.12.006] [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: 09/15/2023] [Revised: 09/30/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025]
Abstract
Embryo development begins with zygotic genome activation (ZGA), eventually generating blastocysts for implantation. However, in vitro systems modeling the pre-implantation development are still absent and challenging. Here, we used mouse totipotent blastomere-like cells (TBLCs) to develop spontaneous differentiation and blastoid formation systems, respectively. We found Wnt signaling enabled the rapid expansion of TBLCs and the optimization of their culture medium. We successfully developed a TBLC-spontaneous differentiation system in which mouse TBLCs (mTBLCs) firstly converted into two types of ZGA-like cells (ZLCs) distinguished by Zscan4 expression. Surprisingly, Zscan4-, but not Zscan4+, ZLCs further passed through intermediate 4-cell and then 8-cell/morula stages to produce epiblast, primitive endoderm, and trophectoderm lineages. Significantly, single TBLCs underwent expansion, compaction, and polarization to efficiently generate blastocyst-like structures and even post-implantation egg-cylinder-like structures. Conclusively, we established TBLC-based differentiation and embryo-like structure formation systems to model early embryonic development, offering criteria for evaluating and understanding totipotency.
Collapse
Affiliation(s)
- Bing Peng
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Qingyi Wang
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Feixiang Zhang
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Hui Shen
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Peng Du
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Beijing Advanced Center of RNA Biology, Peking University, Beijing 100871, China.
| |
Collapse
|
20
|
Wang L, Wang C, Moriano JA, Chen S, Zuo G, Cebrián-Silla A, Zhang S, Mukhtar T, Wang S, Song M, de Oliveira LG, Bi Q, Augustin JJ, Ge X, Paredes MF, Huang EJ, Alvarez-Buylla A, Duan X, Li J, Kriegstein AR. Molecular and cellular dynamics of the developing human neocortex. Nature 2025:10.1038/s41586-024-08351-7. [PMID: 39779846 DOI: 10.1038/s41586-024-08351-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 10/31/2024] [Indexed: 01/11/2025]
Abstract
The development of the human neocortex is highly dynamic, involving complex cellular trajectories controlled by gene regulation1. Here we collected paired single-nucleus chromatin accessibility and transcriptome data from 38 human neocortical samples encompassing both the prefrontal cortex and the primary visual cortex. These samples span five main developmental stages, ranging from the first trimester to adolescence. In parallel, we performed spatial transcriptomic analysis on a subset of the samples to illustrate spatial organization and intercellular communication. This atlas enables us to catalogue cell-type-specific, age-specific and area-specific gene regulatory networks underlying neural differentiation. Moreover, combining single-cell profiling, progenitor purification and lineage-tracing experiments, we have untangled the complex lineage relationships among progenitor subtypes during the neurogenesis-to-gliogenesis transition. We identified a tripotential intermediate progenitor subtype-tripotential intermediate progenitor cells (Tri-IPCs)-that is responsible for the local production of GABAergic neurons, oligodendrocyte precursor cells and astrocytes. Notably, most glioblastoma cells resemble Tri-IPCs at the transcriptomic level, suggesting that cancer cells hijack developmental processes to enhance growth and heterogeneity. Furthermore, by integrating our atlas data with large-scale genome-wide association study data, we created a disease-risk map highlighting enriched risk associated with autism spectrum disorder in second-trimester intratelencephalic neurons. Our study sheds light on the molecular and cellular dynamics of the developing human neocortex.
Collapse
Affiliation(s)
- Li Wang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| | - Cheng Wang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Juan A Moriano
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- University of Barcelona Institute of Complex Systems, Barcelona, Spain
| | - Songcang Chen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Guolong Zuo
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Arantxa Cebrián-Silla
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Shaobo Zhang
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | - Tanzila Mukhtar
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Shaohui Wang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Mengyi Song
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Lilian Gomes de Oliveira
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Neuro-immune Interactions Laboratory, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Qiuli Bi
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jonathan J Augustin
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Xinxin Ge
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Mercedes F Paredes
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Eric J Huang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Arturo Alvarez-Buylla
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Xin Duan
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Jingjing Li
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| | - Arnold R Kriegstein
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Yu Q, Wu Y, Ma X, Zhang Y. Causal genes identification of giant cell arteritis in CD4+ Memory t cells: an integration of multi-omics and expression quantitative trait locus analysis. Inflamm Res 2025; 74:3. [PMID: 39762453 PMCID: PMC11703992 DOI: 10.1007/s00011-024-01965-7] [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: 10/13/2024] [Revised: 10/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Giant cell arteritis (GCA) is a prevalent artery and is strongly correlated with age. The role of CD4+ Memory T cells in giant cell arteritis has not been elucidated. METHOD Through single-cell analysis, we focused on the CD4+ Memory T cells in giant cell arteritis. eQTL analysis and mendelian randomization analysis identified the significant genes which have a causal effect on giant cell arteritis risk. CD4+ Memory T cells were subsequently divided into gene-positive and gene-negative groups, then further single-cell analysis was conducted. Mendelian randomization of plasma proteins, blood-urine biomarkers and metabolites were also performed. Eventually, the PMA induced Jurkat cell lines were used for biological experiments to explore the specific functions of significant causal genes in CD4+ Memory T cells. RESULTS Similarity of CD4+ Memory T cells in GCA and old samples were explored. DDIT4 and ARHGAP15 were identified as significant risk genes via mendelian randomization. The CD4+ Memory T cells were then divided into DDIT4 ± or ARHGAP15 ± groups, and further single-cell analysis indicated the differences in aspects involving intercellular communication, functional pathways, protein activity, metabolism and drug sensitivity between positive and negative groups. In vitro experiments, including overexpression and knockdown, demonstrated that DDIT4 leading to a chronic, low-intensity inflammatory state in CD4+ Memory T cells, eventually promoting the development of GCA. CONCLUSION DDIT4 and ARHGAP15 have significant causal effects on giant cell arteritis risk. Specifically, DDIT4 exhibit pro-inflammatory effects on GCA via promotes chronic, low-intensity inflammatory in CD4+ Memory T cell.
Collapse
Affiliation(s)
- Qiyi Yu
- Carnegie Mellon University, Pittsburgh, USA.
| | - Yifan Wu
- Mudi Meng Honors College, China Pharmaceutical University, Nanjing, China
| | - Xianda Ma
- Carnegie Mellon University, Pittsburgh, USA
| | - Yidong Zhang
- Queen's Belfast University, Belfast, Northern Ireland, UK
| |
Collapse
|
22
|
Muñoz-Blat I, Pérez-Moraga R, Castillo-Marco N, Cordero T, Ochando A, Ortega-Sanchís S, Parras-Moltó M, Monfort-Ortiz R, Satorres-Perez E, Novillo B, Perales A, Gormley M, Granados-Aparici S, Noguera R, Roson B, Fisher SJ, Simón C, Garrido-Gómez T. Multi-omics-based mapping of decidualization resistance in patients with a history of severe preeclampsia. Nat Med 2025:10.1038/s41591-024-03407-7. [PMID: 39775038 DOI: 10.1038/s41591-024-03407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 11/08/2024] [Indexed: 01/11/2025]
Abstract
Endometrial decidualization resistance (DR) is implicated in various gynecological and obstetric conditions. Here, using a multi-omic strategy, we unraveled the cellular and molecular characteristics of DR in patients who have suffered severe preeclampsia (sPE). Morphological analysis unveiled significant glandular anatomical abnormalities, confirmed histologically and quantified by the digitization of hematoxylin and eosin-stained tissue sections. Single-cell RNA sequencing (scRNA-seq) of endometrial samples from patients with sPE (n = 11) and controls (n = 12) revealed sPE-associated shifts in cell composition, manifesting as a stromal mosaic state characterized by proliferative stromal cells (MMP11 and SFRP4) alongside IGFBP1+ decidualized cells, with concurrent epithelial mosaicism and a dearth of epithelial-stromal transition associated with decidualization. Cell-cell communication network mapping underscored aberrant crosstalk among specific cell types, implicating crucial pathways such as endoglin, WNT and SPP1. Spatial transcriptomics in a replication cohort validated DR-associated features. Laser capture microdissection/mass spectrometry in a second replication cohort corroborated several scRNA-seq findings, notably the absence of stromal to epithelial transition at a pathway level, indicating a disrupted response to steroid hormones, particularly estrogens. These insights shed light on potential molecular mechanisms underpinning DR pathogenesis in the context of sPE.
Collapse
Affiliation(s)
- Irene Muñoz-Blat
- Carlos Simon Foundation, Valencia, Spain
- INCLIVA Health Research Institute, Valencia, Spain
| | | | | | | | | | | | | | - Rogelio Monfort-Ortiz
- Department of Obstetrics and Gynecology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Elena Satorres-Perez
- Department of Obstetrics and Gynecology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Blanca Novillo
- Department of Obstetrics and Gynecology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Alfredo Perales
- Department of Obstetrics and Gynecology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Matthew Gormley
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center of Reproductive Science, University of California San Francisco, San Francisco, CA, USA
| | - Sofia Granados-Aparici
- INCLIVA Health Research Institute, Valencia, Spain
- Department of Pathology, Medical School, University of Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Noguera
- INCLIVA Health Research Institute, Valencia, Spain
- Department of Pathology, Medical School, University of Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Susan J Fisher
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center of Reproductive Science, University of California San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Sandler-Moore Mass Spectrometry Core Facility, University of California San Francisco, San Francisco, CA, USA
| | - Carlos Simón
- Carlos Simon Foundation, Valencia, Spain.
- INCLIVA Health Research Institute, Valencia, Spain.
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain.
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Tamara Garrido-Gómez
- Carlos Simon Foundation, Valencia, Spain.
- INCLIVA Health Research Institute, Valencia, Spain.
| |
Collapse
|
23
|
Kaldhusdal V, Boger MF, Tjernlund A, Burgener AD, Bradley F, Lajoie J, Omollo K, Kimani J, Fowke K, Czarnewski P, Broliden K. Spatial transcriptomics unveils estrogen-modulated immune responses and structural alterations in the ectocervical mucosa of depot medroxyprogesterone acetate users. Sci Rep 2025; 15:1014. [PMID: 39762272 PMCID: PMC11704007 DOI: 10.1038/s41598-024-83775-9] [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: 07/11/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
The injectable contraceptive, depot medroxyprogesterone acetate (DMPA), is associated with compromised cervical mucosal barriers. High-resolution spatial transcriptomics is applied here to reveal the spatial localization of these altered molecular markers. Ectocervical tissue samples from Kenyan sex workers using DMPA, or non-hormonal contraceptives, underwent spatial transcriptomics and gene set enrichment analyses. Integrated systemic estradiol levels and bulk tissue gene expression data from a larger cohort enhanced the study's scope. Unsupervised clustering unveiled four epithelial and seven submucosal layers, showcasing spatially restricted and diverse functional epithelial responses, and a less structured submucosal spatial ordering. DMPA associated with mucosal-wide immunoglobulin gene upregulation, verified by CD20+ B-cell immunostaining, and upregulated immune markers adjacent to the basal membrane. Downregulated genes represented spatially restricted disrupted epithelial barrier integrity and submucosal extracellular matrix dysfunction. The transcriptional profile was associated with markers of estrogen regulation. Collectively, our findings reveal estrogen-modulated distinct ectocervical transcriptional profiles associated with DMPA usage. While upregulation of immunoglobulin genes occurs throughout the mucosa, activation of innate immune responses and dysregulation of barrier integrity markers are spatially restricted. These results extend previous analyses using bulk transcriptomics and provide insights into the molecular landscape influenced by DMPA, shedding light on contraceptive effects and health implications.
Collapse
Affiliation(s)
- Vilde Kaldhusdal
- Department of Medicine Solna, Division of Infectious Diseases, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Bioclinicum J7:20, 171 76, Solna, Sweden.
| | - Mathias Franzen Boger
- Department of Medicine Solna, Division of Infectious Diseases, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Bioclinicum J7:20, 171 76, Solna, Sweden
| | - Annelie Tjernlund
- Department of Medicine Solna, Division of Infectious Diseases, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Bioclinicum J7:20, 171 76, Solna, Sweden
| | - Adam D Burgener
- Department of Medicine Solna, Division of Infectious Diseases, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Bioclinicum J7:20, 171 76, Solna, Sweden
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Obstetrics & Gynecology, University of Manitoba, Winnipeg, MB, R3E 3P5, Canada
| | - Frideborg Bradley
- Department of Medicine Solna, Division of Infectious Diseases, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Bioclinicum J7:20, 171 76, Solna, Sweden
| | - Julie Lajoie
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada
- Department of Medical Microbiology and Immunology, University of Nairobi, Kenyatta National Hospital Campus, 30197-00100, Nairobi, Kenya
| | - Kenneth Omollo
- Department of Medical Microbiology and Immunology, University of Nairobi, Kenyatta National Hospital Campus, 30197-00100, Nairobi, Kenya
| | - Joshua Kimani
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada
- Department of Medical Microbiology and Immunology, University of Nairobi, Kenyatta National Hospital Campus, 30197-00100, Nairobi, Kenya
- Partners for Health and Development in Africa, 3737-00506, Nairobi, Kenya
| | - Keith Fowke
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada
- Department of Medical Microbiology and Immunology, University of Nairobi, Kenyatta National Hospital Campus, 30197-00100, Nairobi, Kenya
- Partners for Health and Development in Africa, 3737-00506, Nairobi, Kenya
- Department of Community Health Sciences, University of Manitoba, Winnipeg, MB, R3E 0W3, Canada
| | - Paulo Czarnewski
- Science for Life Laboratory, Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Stockholm University, 171 65, Solna, Sweden
| | - Kristina Broliden
- Department of Medicine Solna, Division of Infectious Diseases, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institutet, Bioclinicum J7:20, 171 76, Solna, Sweden
| |
Collapse
|
24
|
Coulombe P, Tomellini E, Chagraoui J, Mayotte N, Sauvageau G. Deciphering the effect of UM171 on human hematopoietic progenitor cell fate through clonal analysis. Nat Commun 2025; 16:195. [PMID: 39747844 PMCID: PMC11696577 DOI: 10.1038/s41467-024-55225-7] [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: 04/17/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
Ex vivo expansion of hematopoietic stem cells (HSC) requires the maintenance of a stemness state while cells are proliferating. This can be achieved via exposure to UM171 which leads to the degradation of chromatin modifiers and prevents the loss of key epigenetic marks. However, the chromatin landscape varies across populations within the hematopoietic system and the effect of UM171 on self-renewal and differentiation potential of different hematopoietic progenitor cells is less characterized. To address this, we use the CellTag barcoding approach to track the fate of individual stem and progenitor cells during in vitro expansion. We show that, in addition to its HSC self-renewing property, UM171 specifically modulates cell fate of a precursor common to erythroid, megakaryocytic, and mast cells in favor of self-renewal and a mast-bias differentiation trajectory. This differentiation bias can be driven by pro-inflammatory signaling pathways that are activated downstream of UM171 and results in an abundant mast cell population that can be transplanted as part of the graft to populate mice tissues in xenotransplantation studies.
Collapse
Affiliation(s)
- Patrick Coulombe
- Molecular Genetics of Stem Cells Laboratory, Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC, Canada
| | - Elisa Tomellini
- Molecular Genetics of Stem Cells Laboratory, Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC, Canada
| | - Jalila Chagraoui
- Molecular Genetics of Stem Cells Laboratory, Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC, Canada
| | - Nadine Mayotte
- Molecular Genetics of Stem Cells Laboratory, Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC, Canada
| | - Guy Sauvageau
- Molecular Genetics of Stem Cells Laboratory, Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC, Canada.
- Division of Hematology, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada.
- Department of Medicine, Faculty of Medicine, University de Montreal, Montreal, QC, Canada.
| |
Collapse
|
25
|
Araki D, Chen V, Redekar N, Salisbury-Ruf C, Luo Y, Liu P, Li Y, Smith RH, Dagur P, Combs C, Larochelle A. Post-transplant G-CSF impedes engraftment of gene-edited human hematopoietic stem cells by exacerbating p53-mediated DNA damage response. Cell Stem Cell 2025; 32:53-70.e8. [PMID: 39536761 PMCID: PMC11698648 DOI: 10.1016/j.stem.2024.10.013] [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: 07/06/2023] [Revised: 05/06/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Granulocyte-colony-stimulating factor (G-CSF) is commonly used to accelerate recovery from neutropenia following chemotherapy and autologous transplantation of hematopoietic stem and progenitor cells (HSPCs) for malignant disorders. However, its utility after ex vivo gene therapy in human HSPCs remains unexplored. We show that administering G-CSF from day 1 to 14 post-transplant impedes engraftment of CRISPR-Cas9 gene-edited human HSPCs in murine xenograft models. G-CSF affects gene-edited HSPCs through a cell-intrinsic mechanism, causing proliferative stress and amplifying the early p53-mediated DNA damage response triggered by Cas9-mediated DNA double-strand breaks. This underscores a threshold mechanism where p53 activation must reach a critical level to impair cellular function. Transiently inhibiting p53 or delaying the initiation of G-CSF treatment to day 5 post-transplant attenuates its negative impact on gene-edited HSPCs. The potential for increased HSPC toxicity associated with post-transplant G-CSF administration in CRISPR-Cas9 autologous HSPC gene therapy warrants consideration in clinical trials.
Collapse
Affiliation(s)
- Daisuke Araki
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Vicky Chen
- Integrated Data Science Services (IDSS), National Institutes of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA
| | - Neelam Redekar
- Integrated Data Science Services (IDSS), National Institutes of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA
| | - Christi Salisbury-Ruf
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Yan Luo
- DNA Sequencing and Genomics Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Poching Liu
- DNA Sequencing and Genomics Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Yuesheng Li
- DNA Sequencing and Genomics Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Richard H Smith
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Pradeep Dagur
- Flow Cytometry Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Christian Combs
- Light Microscopy Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Andre Larochelle
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
26
|
Li J, Tan LX, Sun B, Griffin N, Niknezhad SV, Yu C, Berthoin L, Cruz-Pacheco N, Mohabbat S, Sinada H, Efraim Y, Ting Chen FY, An L, Gaylord EA, Bahney CS, Lombaert IM, Knox SM. Chronic degenerative failure of salivary glands can be reversed through restoring mitochondrial function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630834. [PMID: 39803569 PMCID: PMC11722244 DOI: 10.1101/2024.12.31.630834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
There are no therapies for reversing chronic organ degeneration. Non-healing degenerative wounds are thought to be irreparable, in part, by the inability of the tissue to respond to reparative stimuli. As such, treatments are typically aimed at slowing tissue degeneration or replacing cells through transplantation. Building on our previous studies showing acutely injured salivary glands, and specifically secretory acini, can be regenerated, we reveal that non-healing, degenerating murine salivary glands remain responsive to a neuromimetic (muscarinic) agonist with treatment resulting in the restoration of tissue. Not only is degenerated tissue structure and function returned to a homeostatic-like state, but this outcome is also sustained months after treatment termination. Furthermore, despite an eventual reduction in saliva secretion, the gland responds to a second round of treatment, fully regaining secretory function that resembles uninjured controls. Our findings suggest this rescue is due to a reversal of an aberrant de-differentiated acinar cell state and mitochondrial dysfunction through a muscarinic-calcium signaling pathway. Thus, these data challenge the concept that organ degeneration is irreversible and provides a readily testable therapeutic strategy for epithelial organ restoration that may significantly benefit a diversity of chronic disease conditions.
Collapse
Affiliation(s)
- Jianlong Li
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
- These authors contributed equally
| | - Li Xuan Tan
- Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, California, USA; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
- These authors contributed equally
| | - Bo Sun
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
- These authors contributed equally
| | - Nathan Griffin
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Seyyed Vahid Niknezhad
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Chieh Yu
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Lionel Berthoin
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Noel Cruz-Pacheco
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Seayar Mohabbat
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Hanan Sinada
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Yael Efraim
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Feeling Yu Ting Chen
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Luye An
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Eliza A. Gaylord
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Chelsey S. Bahney
- University of California, San Francisco. Orthopedic Trauma Institute, San Francisco, CA
| | - Isabelle M.A. Lombaert
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA
- Co–senior authors
| | - Sarah M. Knox
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, California, USA
- Co–senior authors
- Lead contact
| |
Collapse
|
27
|
Deng K, Xu X, Zhou M, Li H, Keller ET, Shelley G, Lu A, Garmire L, Guan Y. ImageDoubler: image-based doublet identification in single-cell sequencing. Nat Commun 2025; 16:21. [PMID: 39747095 PMCID: PMC11695948 DOI: 10.1038/s41467-024-55434-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Single-cell sequencing provides detailed insights into individual cell behaviors within complex systems based on the assumption that each cell is uniquely isolated. However, doublets-where two or more cells are sequenced together-disrupt this assumption and can lead to potential data misinterpretations. Traditional doublet detection methods primarily rely on simulated genomic data, which may be less effective in homogeneous cell populations and can introduce biases from experimental processes. Therefore, we introduce ImageDoubler in this study, an innovative image-based model that identifies doublets and missing samples leveraging the Fluidigm single-cell sequencing image data. Our approach showcases a notable doublet detection efficacy, achieving a rate up to 93.87% and registering a minimum improvement of 33.1% in F1 scores compared to existing genomic-based methods. This advancement highlights the potential of using imaging to glean insight into developing doublet detection algorithms and exposes the limitations inherent in current genomic-based techniques.
Collapse
Affiliation(s)
- Kaiwen Deng
- Gilbert S. Omenn Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Xinya Xu
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Manqi Zhou
- Gilbert S. Omenn Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Hongyang Li
- Gilbert S. Omenn Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Evan T Keller
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Gregory Shelley
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Annie Lu
- Gilbert S. Omenn Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Lana Garmire
- Gilbert S. Omenn Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Yuanfang Guan
- Gilbert S. Omenn Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
28
|
Holmes AB, Corinaldesi C, Basso K. Single-Cell Transcriptomic Analysis of Normal and Malignant B Cells. Methods Mol Biol 2025; 2865:347-374. [PMID: 39424732 DOI: 10.1007/978-1-0716-4188-0_15] [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: 10/21/2024]
Abstract
In the past decade, single-cell (sc) transcriptomics has overcome the limitations of bulk analysis by measuring gene expression in individual cells, not just a population average. This can identify diverse cell types and states within a sample with high resolution, even without prior purification. Various technologies exist, each with its own capture, barcoding, and library preparation methods. This chapter focuses on the analysis of normal and malignant mature B cells using the 10× Genomics 5' sc-gene expression in parallel with B cell immune repertoire profiling. By integrating the gene expression data from similar cells, the complete transcriptome for each population can be reconstructed, while the identification of the expressed immunoglobulin genes allows investigating clonotype evolution and the detection of tumor clones that share the same clonally rearranged B cell receptor sequence. Researchers are guided through both the experimental protocols and data analysis with a comprehensive, step-by-step walkthrough of how to use some of the more popular single-cell software tools.
Collapse
Affiliation(s)
- Antony B Holmes
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | | | - Katia Basso
- Institute for Cancer Genetics and Department of Pathology & Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
29
|
Wu J, Xia S, Ye W, Sun Y, Cai J, Yu F, Wen H, Yi X, Li T, Chen M, Chen J, Song G, Yang C, Song Y, Wang J. Dissecting the cell microenvironment of ovarian endometrioma through single-cell RNA sequencing. SCIENCE CHINA. LIFE SCIENCES 2025; 68:116-129. [PMID: 39470923 DOI: 10.1007/s11427-024-2638-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/16/2024] [Indexed: 11/01/2024]
Abstract
Ovarian endometrioma (OE), also known as "chocolate cysts," is a cystic mass that develops in the ovaries due to endometriosis and is a common gynecological condition characterized by the growth of endometrial tissue outside the uterus, leading to symptoms such as dysmenorrhea, pelvic pain, and infertility. However, the precise molecular and cellular mechanisms driving this pathophysiology remain largely unknown, posing challenges for diagnosis and treatment. Here, we employed integrated single-cell transcriptomic profiling of over 52,000 individual cells from endometrial tissues of OE patients and healthy donors and identified twelve major cell populations. We identified notable alterations in cell type-specific proportions and molecular signatures associated with OE. Notably, the activation of IGFBP5+ macrophages with pro-inflammatory properties, NK cell exhaustion, and aberrant proliferation of IQCG+ and KLF2+ epithelium are key features and may be the potential mechanisms underlying the pathogenesis of OE. Collectively, our data contribute to a better understanding of OE at the single cell level and may pave the way for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Jiangpeng Wu
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China
- Department of Urology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Southern University of Science and Technology, Shenzhen, 518020, China
| | - Siyu Xia
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China
| | - Wenting Ye
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China
| | - Yan Sun
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China
| | - Jing Cai
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China
| | - Fubing Yu
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China
| | - Haiping Wen
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China
| | - Xiuwei Yi
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China
| | - Taikang Li
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China
| | - Mingwei Chen
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China
| | - Jiayun Chen
- Department of Urology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Southern University of Science and Technology, Shenzhen, 518020, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ge Song
- Reproductive Medicine Center, Foshan Women and Children Hospital, Foshan, 528000, China.
| | - Chuanbin Yang
- Department of Urology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Southern University of Science and Technology, Shenzhen, 518020, China.
| | - Yali Song
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China.
| | - Jigang Wang
- Department of Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Dongguan, 523000, China.
- Department of Urology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Southern University of Science and Technology, Shenzhen, 518020, China.
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
30
|
Yin M, Sun L, Wu S, Ma J, Zhang W, Ji X, Tang Z, Zhang X, Yang Y, Zhang X, Huang J, Zheng S, Liu W, Ji C, Zhang L. TGFβ-mediated inhibition of hypodermal adipocyte progenitor differentiation promotes wound-induced skin fibrosis. Cell Prolif 2025; 58:e13722. [PMID: 39072821 PMCID: PMC11693572 DOI: 10.1111/cpr.13722] [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: 03/25/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
Aberrant activation of dermal fibroblasts during wound healing often leads to debilitating fibrotic changes in the skin, such as scleroderma and keloids. However, the underlying cellular and molecular mechanisms remain elusive. Here, we established a wound-induced skin fibrosis (WISF) mouse model in mature adult mice, characterised by excessive deposition of collagen bundles, loss of dermal adipocytes, and enrichment of DPP4+Ly6A+THY1+ hypodermal interstitial adipocyte progenitors (HI-APs) and pericytes, resembling human fibrotic skin diseases. This WISF model exhibited an age-dependent gain of fibrotic characteristics, contrasting with the wound-induced hair neogenesis observed in younger mice. Through comprehensive analyses of the WISF, we delineated a trajectory of fibroblast differentiation that originates from HI-APs. These progenitors highly expressed several extracellular matrix (ECM) genes and exhibited a TGFβ pathway signature. TGFβ was identified as the key signal to inhibit the adipogenic potential and maintain the fibrogenic potential of dermal APs. Additionally, administering a TGFβ receptor inhibitor to wound scar reduced the abundance of ECM-producing APs. Finally, analysis of human scleroderma skin tissues revealed a negative correlation between the expression of AP-, ECM-, and TGFβ pathway-related genes and PPARG. Overall, this study establishes a wound-induced skin fibrosis mouse model and demonstrates that TGFβ-mediated blockage of HI-AP differentiation is crucial for driving fibrotic pathology. Targeting HI-APs and adipogenesis may provide novel avenues for developing disease-modifying therapies for fibrotic skin diseases.
Collapse
Affiliation(s)
- Meimei Yin
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical SciencesXiamen UniversityXiamenChina
| | - Lixiang Sun
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical SciencesXiamen UniversityXiamenChina
- Central Laboratory, Zhongshan Hospital (Xiamen)Fudan UniversityXiamenChina
| | - Shuai Wu
- State Key Laboratory of Cellular Stress Biology, School of Life SciencesXiamen UniversityXiamenChina
| | - Jinhang Ma
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical SciencesXiamen UniversityXiamenChina
| | - Wenlu Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical SciencesXiamen UniversityXiamenChina
| | - Xiaoxuan Ji
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical SciencesXiamen UniversityXiamenChina
| | - Zhichong Tang
- State Key Laboratory of Cellular Stress Biology, School of Life SciencesXiamen UniversityXiamenChina
| | - Xiaowei Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical SciencesXiamen UniversityXiamenChina
| | - Yichun Yang
- Central Laboratory, Zhongshan Hospital (Xiamen)Fudan UniversityXiamenChina
| | - Xinyuan Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical SciencesXiamen UniversityXiamenChina
| | - Jin‐wen Huang
- Department of DermatologyThe First Affiliated Hospital of Fujian Medical UniversityFuzhouChina
| | - Shaoluan Zheng
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital (Xiamen)Fudan UniversityXiamenChina
| | - Wen‐jie Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical SciencesXiamen UniversityXiamenChina
| | - Chao Ji
- Department of DermatologyThe First Affiliated Hospital of Fujian Medical UniversityFuzhouChina
| | - Ling‐juan Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical SciencesXiamen UniversityXiamenChina
| |
Collapse
|
31
|
Huang QF, Wang GF, Zhang YM, Zhang C, Ran YQ, He JZ, Wang G, Xu XE, Wang SH, Wu JY, Li EM, Xu LY. Lympho-myeloid aggregate-infiltrating CD20 + B cells display a double-negative phenotype and correlate with poor prognosis in esophageal squamous cell carcinoma. Transl Res 2025; 275:48-61. [PMID: 39536938 DOI: 10.1016/j.trsl.2024.11.002] [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: 04/11/2024] [Revised: 09/19/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
According to morphological features, tumor-infiltrating B cells (TIL-Bs) can be classified as lympho-myeloid aggregates (LMAs) and tertiary lymphoid structures (TLSs). As a disease with high incidence and mortality, research on esophageal squamous cell carcinoma (ESCC) TIL-Bs is still unclear. Thus, we aimed to investigate the prognostic value and functional involvement of TIL-Bs in ESCC. Based on CD20 immunohistochemical staining of 147 ESCC samples, the TIL-Bs at different anatomic subregions (intra-tumor (T), invasive margin (IM) and peri-tumor (P)) were quantified and correlated with survival by Kaplan-Meier analyses. We found that LMAs were widely distributed throughout the whole section and were associated with poor prognosis, especially those located in the T subregion, which was contrary to the positive clinical significance of TLSs. Based on the number of LMAs and TLSs, a four-level immune type was constructed as an independent predictor for survival. Using multiplexed immunofluorescence (mIF) staining, we found that the main phenotype of infiltrating B cells in LMAs was CD20+IgD-CD27- double-negative (DN) B cells. DN B cells were abundant in ESCC tumor tissue, and their high expression was related to shortened overall survival time. Subsequently, we demonstrate a close relationship between DN B cells and regulatory T cells (Tregs) using single cell RNA-seq data, bulk RNA-seq data and flow cytometry, and verified the spatial proximity of DN B cells and Tregs by mIF staining. Trajectory analysis and flow cytometry revealed that DN B cells highly expressed genes involved in the antigen processing and presentation pathway, such as HLA-DR. The abundance of DN B cells and LMAs in ESCC provides novel potential targets for optimal immunotherapy against ESCC.
Collapse
Affiliation(s)
- Qing-Feng Huang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Ge-Fei Wang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Yi-Meng Zhang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Cong Zhang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Ying-Qi Ran
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Jian-Zhong He
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, PR China
| | - Geng Wang
- Department of Thoracic Surgery, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Xiu-E Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - Shao-Hong Wang
- Departments of Pathology, Shantou Central Hospital, Shantou 515041, Guangdong, PR China
| | - Jian-Yi Wu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China
| | - En-Min Li
- Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Shantou Academy Medical Sciences, Shantou 515041, Guangdong, PR China; Guangdong Esophageal Cancer Research Institute, Shantou Sub-center, Cancer Research Center, Shantou University Medical College, Shantou 515041, Guangdong, PR China.
| | - Li-Yan Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, PR China; Guangdong Esophageal Cancer Research Institute, Shantou Sub-center, Cancer Research Center, Shantou University Medical College, Shantou 515041, Guangdong, PR China.
| |
Collapse
|
32
|
Hamed AA, Hua K, Trinh QM, Simons BD, Marioni JC, Stein LD, Dirks PB. Gliomagenesis mimics an injury response orchestrated by neural crest-like cells. Nature 2025:10.1038/s41586-024-08356-2. [PMID: 39743595 DOI: 10.1038/s41586-024-08356-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
Glioblastoma is an incurable brain malignancy. By the time of clinical diagnosis, these tumours exhibit a degree of genetic and cellular heterogeneity that provides few clues to the mechanisms that initiate and drive gliomagenesis1,2. Here, to explore the early steps in gliomagenesis, we utilized conditional gene deletion and lineage tracing in tumour mouse models, coupled with serial magnetic resonance imaging, to initiate and then closely track tumour formation. We isolated labelled and unlabelled cells at multiple stages-before the first visible abnormality, at the time of the first visible lesion, and then through the stages of tumour growth-and subjected cells of each stage to single-cell profiling. We identify a malignant cell state with a neural crest-like gene expression signature that is highly abundant in the early stages, but relatively diminished in the late stage of tumour growth. Genomic analysis based on the presence of copy number alterations suggests that these neural crest-like states exist as part of a heterogeneous clonal hierarchy that evolves with tumour growth. By exploring the injury response in wounded normal mouse brains, we identify cells with a similar signature that emerge following injury and then disappear over time, suggesting that activation of an injury response program occurs during tumorigenesis. Indeed, our experiments reveal a non-malignant injury-like microenvironment that is initiated in the brain following oncogene activation in cerebral precursor cells. Collectively, our findings provide insight into the early stages of glioblastoma, identifying a unique cell state and an injury response program tied to early tumour formation. These findings have implications for glioblastoma therapies and raise new possibilities for early diagnosis and prevention of disease.
Collapse
Affiliation(s)
- Akram A Hamed
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Developmental and Stem Cell Biology Department, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kui Hua
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Quang M Trinh
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Benjamin D Simons
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK.
- Wellcome Sanger Institute, Cambridge, UK.
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, Cambridge, UK.
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK.
| | - John C Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK.
- Wellcome Sanger Institute, Cambridge, UK.
| | - Lincoln D Stein
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada.
| | - Peter B Dirks
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
- Developmental and Stem Cell Biology Department, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
33
|
Rito T, Libby ARG, Demuth M, Domart MC, Cornwall-Scoones J, Briscoe J. Timely TGFβ signalling inhibition induces notochord. Nature 2025; 637:673-682. [PMID: 39695233 PMCID: PMC11735409 DOI: 10.1038/s41586-024-08332-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: 02/27/2023] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
The formation of the vertebrate body involves the coordinated production of trunk tissues from progenitors located in the posterior of the embryo. Although in vitro models using pluripotent stem cells replicate aspects of this process1-10, they lack crucial components, most notably the notochord-a defining feature of chordates that patterns surrounding tissues11. Consequently, cell types dependent on notochord signals are absent from current models of human trunk formation. Here we performed single-cell transcriptomic analysis of chick embryos to map molecularly distinct progenitor populations and their spatial organization. Guided by this map, we investigated how differentiating human pluripotent stem cells develop a stereotypical spatial organization of trunk cell types. We found that YAP inactivation in conjunction with FGF-mediated MAPK signalling facilitated WNT pathway activation and induced expression of TBXT (also known as BRA). In addition, timely inhibition of WNT-induced NODAL and BMP signalling regulated the proportions of different tissue types, including notochordal cells. This enabled us to create a three-dimensional model of human trunk development that undergoes morphogenetic movements, producing elongated structures with a notochord and ventral neural and mesodermal tissues. Our findings provide insights into the mechanisms underlying vertebrate notochord formation and establish a more comprehensive in vitro model of human trunk development. This paves the way for future studies of tissue patterning in a physiologically relevant environment.
Collapse
Affiliation(s)
- Tiago Rito
- The Francis Crick Institute, London, UK.
| | | | | | | | | | | |
Collapse
|
34
|
Li F, Dai P, Shi H, Zhang Y, He J, Gopalan A, Li D, Chen Y, Du Y, Xu G, Yang W, Liang C, Gao D. LKB1 inactivation promotes epigenetic remodeling-induced lineage plasticity and antiandrogen resistance in prostate cancer. Cell Res 2025; 35:59-71. [PMID: 39743630 DOI: 10.1038/s41422-024-01025-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/22/2024] [Indexed: 01/04/2025] Open
Abstract
Epigenetic regulation profoundly influences the fate of cancer cells and their capacity to switch between lineages by modulating essential gene expression, thereby shaping tumor heterogeneity and therapy response. In castration-resistant prostate cancer (CRPC), the intricacies behind androgen receptor (AR)-independent lineage plasticity remain unclear, leading to a scarcity of effective clinical treatments. Utilizing single-cell RNA sequencing on both human and mouse prostate cancer samples, combined with whole-genome bisulfite sequencing and multiple genetically engineered mouse models, we investigated the molecular mechanism of AR-independent lineage plasticity and uncovered a potential therapeutic strategy. Single-cell transcriptomic profiling of human prostate cancers, both pre- and post-androgen deprivation therapy, revealed an association between liver kinase B1 (LKB1) pathway inactivation and AR independence. LKB1 inactivation led to AR-independent lineage plasticity and global DNA hypomethylation during prostate cancer progression. Importantly, the pharmacological inhibition of TET enzymes and supplementation with S-adenosyl methionine were found to effectively suppress AR-independent prostate cancer growth. These insights shed light on the mechanism driving AR-independent lineage plasticity and propose a potential therapeutic strategy by targeting DNA hypomethylation in AR-independent CRPC.
Collapse
Affiliation(s)
- Fei Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Pengfei Dai
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Huili Shi
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yajuan Zhang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan He
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Anuradha Gopalan
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dan Li
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yarui Du
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Guoliang Xu
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Weiwei Yang
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Chao Liang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China.
| |
Collapse
|
35
|
Yu Z, Yang Y, Chen X, Wong K, Zhang Z, Zhao Y, Li X. Accurate Spatial Heterogeneity Dissection and Gene Regulation Interpretation for Spatial Transcriptomics using Dual Graph Contrastive Learning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410081. [PMID: 39605202 PMCID: PMC11744562 DOI: 10.1002/advs.202410081] [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: 08/22/2024] [Revised: 10/31/2024] [Indexed: 11/29/2024]
Abstract
Recent advances in spatial transcriptomics have enabled simultaneous preservation of high-throughput gene expression profiles and the spatial context, enabling high-resolution exploration of distinct regional characterization in tissue. To effectively understand the underlying biological mechanisms within tissue microenvironments, there is a requisite for methods that can accurately capture external spatial heterogeneity and interpret internal gene regulation from spatial transcriptomics data. However, current methods for region identification often lack the simultaneous characterizing of spatial structure and gene regulation, thereby limiting the ability of spatial dissection and gene interpretation. Here, stDCL is developed, a dual graph contrastive learning method to identify spatial domains and interpret gene regulation in spatial transcriptomics data. stDCL adaptively incorporates gene expression data and spatial information via a graph embedding autoencoder, thereby preserving critical information within the latent embedding representations. In addition, dual graph contrastive learning is proposed to train the model, ensuring that the latent embedding representation closely resembles the actual spatial distribution and exhibits cluster similarity. Benchmarking stDCL against other state-of-the-art clustering methods using complex cortex datasets demonstrates its superior accuracy and effectiveness in identifying spatial domains. Our analysis of the imputation matrices generated by stDCL reveals its capability to reconstruct spatial hierarchical structures and refine differential expression assessment. Furthermore, it is demonstrated that the versatility of stDCL in interpretability of gene regulation, spatial heterogeneity at high resolution, and embryonic developmental patterns. In addition, it is also showed that stDCL can successfully annotate disease-associated astrocyte subtypes in Alzheimer's disease and unravel multiple relevant pathways and regulatory mechanisms.
Collapse
Affiliation(s)
- Zhuohan Yu
- School of Artificial IntelligenceJilin UniversityJilin130012China
| | - Yuning Yang
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONM5S 3E1Canada
| | - Xingjian Chen
- Cutaneous Biology Research Center, Massachusetts General HospitalHarvard Medical SchoolBostonMA02115USA
| | - Ka‐Chun Wong
- Department of Computer ScienceCity University of Hong KongHong KongSAR999077Hong Kong
| | - Zhaolei Zhang
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONM5S 3E1Canada
| | - Yuming Zhao
- College of Computer and Control EngineeringNortheast Forestry UniversityHarbin150040China
| | - Xiangtao Li
- School of Artificial IntelligenceJilin UniversityJilin130012China
- Department of Computer ScienceCity University of Hong KongHong KongSAR999077Hong Kong
| |
Collapse
|
36
|
Hou M, Zhao Z, Li S, Zhang Z, Li X, Zhang Y, Huang W, Li L, Xi W, Liang F, Lin L, Zhang Y, Chai G. Single-cell analysis unveils cell subtypes of acral melanoma cells at the early and late differentiation stages. J Cancer 2025; 16:898-916. [PMID: 39781353 PMCID: PMC11705046 DOI: 10.7150/jca.102045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/01/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Melanoma, a malignant neoplasm originating from melanocytes, is a form of skin cancer with rapidly increasing global incidence, often exacerbated by UV radiation[1]. Particularly, acral melanoma, characterized by its swift metastasis and poor prognosis, underscores the significance of further research into its heterogeneity. Single-cell sequencing has been widely utilized in the study of tumor heterogeneity; however, research related to melanoma remains to be further refined. MATERIALS AND METHODS We employed single-cell RNA sequencing (scRNA-seq) transcriptomic analysis to delve into the melanoma cells from six samples of melanoma patients. This approach enabled the identification of critical melanoma cell subpopulations and their roles in melanoma progression. Subsequently, we examined the interactions among these subpopulations and analyzed their interactions with other cell types. RESULTS Our analysis identified C3 ID2+ melanoma cells as an early-stage subpopulation and C4 PCLAF+ cells as a late-stage subpopulation in melanoma evolution. Through our analysis, we identified C4 PCLAF+ Melanoma cells as a significant subpopulation in acral melanoma (AM), playing a pivotal role in the differentiation and development of AM. Further analysis of transcription factors, enriched pathways, cell stemness, and cell trajectories highlighted the significant role of C4 PCLAF+ melanoma cells in acral melanoma (AM) proliferation. CONCLUSION This study identifies new factors influencing melanoma progression, providing a foundation for subsequent research.
Collapse
Affiliation(s)
- Mengyuan Hou
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Zhijie Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Shuxiao Li
- Department of Burns and Plastic Reconstructive Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Province, China
- Life Science and Clinical Medicine Research Center, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Province, China
| | - Ziwei Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Xin Li
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Yichi Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Wenyi Huang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Li Li
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Wenjing Xi
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Feiteng Liang
- Department of Burns and Plastic Reconstructive Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Province, China
- Life Science and Clinical Medicine Research Center, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Province, China
| | - Li Lin
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Yan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| | - Gang Chai
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhi Zao Ju Rd, Shanghai, 200011, China
| |
Collapse
|
37
|
Defard T, Desrentes A, Fouillade C, Mueller F. Homebuilt Imaging-Based Spatial Transcriptomics: Tertiary Lymphoid Structures as a Case Example. Methods Mol Biol 2025; 2864:77-105. [PMID: 39527218 DOI: 10.1007/978-1-0716-4184-2_5] [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: 11/16/2024]
Abstract
Spatial transcriptomics methods provide insight into the cellular heterogeneity and spatial architecture of complex, multicellular systems. Combining molecular and spatial information provides important clues to study tissue architecture in development and disease. Here, we present a comprehensive do-it-yourself (DIY) guide to perform such experiments at reduced costs leveraging open-source approaches. This guide spans the entire life cycle of a project, from its initial definition to experimental choices, wet lab approaches, instrumentation, and analysis. As a concrete example, we focus on tertiary lymphoid structures (TLS), which we use to develop typical questions that can be addressed by these approaches.
Collapse
Affiliation(s)
- Thomas Defard
- Institut Pasteur, Université Paris Cité, Photonic Bio-Imaging, Centre de Ressources et Recherches Technologiques (UTechS-PBI, C2RT), Paris, France
- Institut Pasteur, Université Paris Cité, Imaging and Modeling Unit, Paris, France
- Centre for Computational Biology (CBIO), Mines Paris, PSL University, Paris, France
- Institut Curie, PSL University, Paris, France
- INSERM, U900, Paris, France
| | - Auxence Desrentes
- UMRS1135 Sorbonne University, Paris, France
- INSERM U1135, Paris, France
- Team "Immune Microenvironment and Immunotherapy", Centre for Immunology and Microbial Infections (CIMI), Paris, France
| | - Charles Fouillade
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL Research University, Centre Universitaire, Orsay, France
| | - Florian Mueller
- Institut Pasteur, Université Paris Cité, Photonic Bio-Imaging, Centre de Ressources et Recherches Technologiques (UTechS-PBI, C2RT), Paris, France.
- Institut Pasteur, Université Paris Cité, Imaging and Modeling Unit, Paris, France.
| |
Collapse
|
38
|
Becker TJ, Enkhmandakh B, Bayarsaihan D. Single-cell RNA analysis of chromodomain-encoding genes in mesenchymal stromal cells of the mouse dental pulp. J Cell Biochem 2025; 126:e30608. [PMID: 38779967 DOI: 10.1002/jcb.30608] [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: 10/24/2023] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
The chromodomain helicase DNA-binding (CHD) and chromobox (CBX) families of proteins play crucial roles in cell fate decisions, differentiation, and cell proliferation in a broad variety of tissues and cell types. CHD proteins are ATP-dependent epigenetic enzymes actively engaged in transcriptional regulation, DNA replication, and DNA damage repair, whereas CBX proteins are transcriptional repressors mainly involved in the formation of heterochromatin. The pleiotropic effects of CHD and CBX proteins are largely dependent on their versatility to interact with other key components of the epigenetic and transcriptional machinery. Although the function and regulatory modes of CHD and CBX factors are well established in many cell types, little is known about their roles during osteogenic differentiation. A single-cell RNA-sequencing analysis of the mouse incisor dental pulp revealed distinct spatiotemporal expression patterns of CHD- and CBX-encoding genes within different clusters of mesenchymal stromal cells (MSCs) representing various stages of osteogenic differentiation. Additionally, genes encoding interaction partners of CHD and CBX proteins, such as subunits of the trithorax-COMPASS and polycomb chromatin remodeling complexes, exhibited differential co-expression behaviors within MSC subpopulations. Thus, CHD- and CBX-encoding genes show partially overlapping but distinct expression patterns in MSCs, suggesting their differential roles in osteogenic cell fate decisions.
Collapse
Affiliation(s)
- Timothy James Becker
- Department of Computer Science, Connecticut College, New London, Connecticut, USA
| | - Badam Enkhmandakh
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Dashzeveg Bayarsaihan
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health Center, Farmington, Connecticut, USA
- Institute for System Genomics, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
39
|
Gulati GS, D'Silva JP, Liu Y, Wang L, Newman AM. Profiling cell identity and tissue architecture with single-cell and spatial transcriptomics. Nat Rev Mol Cell Biol 2025; 26:11-31. [PMID: 39169166 DOI: 10.1038/s41580-024-00768-2] [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] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Single-cell transcriptomics has broadened our understanding of cellular diversity and gene expression dynamics in healthy and diseased tissues. Recently, spatial transcriptomics has emerged as a tool to contextualize single cells in multicellular neighbourhoods and to identify spatially recurrent phenotypes, or ecotypes. These technologies have generated vast datasets with targeted-transcriptome and whole-transcriptome profiles of hundreds to millions of cells. Such data have provided new insights into developmental hierarchies, cellular plasticity and diverse tissue microenvironments, and spurred a burst of innovation in computational methods for single-cell analysis. In this Review, we discuss recent advancements, ongoing challenges and prospects in identifying and characterizing cell states and multicellular neighbourhoods. We discuss recent progress in sample processing, data integration, identification of subtle cell states, trajectory modelling, deconvolution and spatial analysis. Furthermore, we discuss the increasing application of deep learning, including foundation models, in analysing single-cell and spatial transcriptomics data. Finally, we discuss recent applications of these tools in the fields of stem cell biology, immunology, and tumour biology, and the future of single-cell and spatial transcriptomics in biological research and its translation to the clinic.
Collapse
Affiliation(s)
- Gunsagar S Gulati
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Yunhe Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Aaron M Newman
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
40
|
Zhao C, Plaza Reyes A, Schell JP, Weltner J, Ortega NM, Zheng Y, Björklund ÅK, Baqué-Vidal L, Sokka J, Trokovic R, Cox B, Rossant J, Fu J, Petropoulos S, Lanner F. A comprehensive human embryo reference tool using single-cell RNA-sequencing data. Nat Methods 2025; 22:193-206. [PMID: 39543283 PMCID: PMC11725501 DOI: 10.1038/s41592-024-02493-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 09/30/2024] [Indexed: 11/17/2024]
Abstract
Stem cell-based embryo models offer unprecedented experimental tools for studying early human development. The usefulness of embryo models hinges on their molecular, cellular and structural fidelities to their in vivo counterparts. To authenticate human embryo models, single-cell RNA sequencing has been utilized for unbiased transcriptional profiling. However, an organized and integrated human single-cell RNA-sequencing dataset, serving as a universal reference for benchmarking human embryo models, remains unavailable. Here we developed such a reference through the integration of six published human datasets covering development from the zygote to the gastrula. Lineage annotations are contrasted and validated with available human and nonhuman primate datasets. Using stabilized Uniform Manifold Approximation and Projection, we constructed an early embryogenesis prediction tool, where query datasets can be projected on the reference and annotated with predicted cell identities. Using this reference tool, we examined published human embryo models, highlighting the risk of misannotation when relevant references are not utilized for benchmarking and authentication.
Collapse
Affiliation(s)
- Cheng Zhao
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Alvaro Plaza Reyes
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
- Department of Integrative Pathophysiology and Therapy, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Seville, Spain
| | - John Paul Schell
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Jere Weltner
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Nicolás M Ortega
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Yi Zheng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA
| | - Åsa K Björklund
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Laura Baqué-Vidal
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Joonas Sokka
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Ras Trokovic
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Brian Cox
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Sophie Petropoulos
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden.
- Département de Médecine, Université de Montréal, Montreal, Quebec, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Axe Immunopathologie, Montreal, Quebec, Canada.
| | - Fredrik Lanner
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden.
- Ming Wai Lau Center for Reparative Medicine, Stockholm Node, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
41
|
Gu X, Kang H, Cao S, Tong Z, Song N. Blockade of TREM2 ameliorates pulmonary inflammation and fibrosis by modulating sphingolipid metabolism. Transl Res 2025; 275:1-17. [PMID: 39490681 DOI: 10.1016/j.trsl.2024.10.002] [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: 01/10/2024] [Revised: 09/19/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Pulmonary fibrosis is a chronic interstitial lung disease involving systemic inflammation and abnormal collagen deposition. Dysregulations in lipid metabolism, such as macrophage-dependent lipid catabolism, have been recognized as critical factors for the development of pulmonary fibrosis. However, little is known about the signaling pathways involved and the key regulators. Here we found that triggering receptor expressed on myeloid cells 2 (TREM2) plays a pivotal role in regulating the lipid handling capacities of pulmonary macrophages and triggering fibrosis. By integrating analysis of single-cell and bulk RNA sequencing data from patients and mice with pulmonary fibrosis, we revealed that pulmonary macrophages consist of heterogeneous populations with distinct pro-fibrotic properties, and found that both sphingolipid metabolism and the expression of chemotaxis-related genes are elevated in fibrotic lungs. TREM2, a sensor recognizing multiple lipid species, is specifically upregulated in a subset of monocyte-derived macrophages. Blockade of TREM2 by conventional/conditional knock-out or soluble TREM2 administration can attenuate bleomycin-induced pulmonary fibrosis. By utilizing scRNA Seq and lipidomics, we found that Trem2 deficiency downregulates the synthesis of various sphingomyelins, and inhibits the expression of chemokines such as Ccl2. Together, our findings not only reveal the alterations in lipidomic profiles and the atlas of pulmonary macrophages during pulmonary fibrosis, but also suggest that targeting TREM2, the crucial regulator affecting both pulmonary sphingolipid metabolism and the chemokines secretion, can benefit pulmonary fibrosis patients in the future.
Collapse
Affiliation(s)
- Xueqing Gu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100020, China
| | - Hanyujie Kang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100020, China
| | - Siyu Cao
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100020, China
| | - Zhaohui Tong
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100020, China.
| | - Nan Song
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100020, China; Medical Research Center, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
42
|
Fang M, Gorin G, Pachter L. Trajectory inference from single-cell genomics data with a process time model. PLoS Comput Biol 2025; 21:e1012752. [PMID: 39836699 PMCID: PMC11760028 DOI: 10.1371/journal.pcbi.1012752] [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: 06/18/2024] [Revised: 01/24/2025] [Accepted: 12/25/2024] [Indexed: 01/23/2025] Open
Abstract
Single-cell transcriptomics experiments provide gene expression snapshots of heterogeneous cell populations across cell states. These snapshots have been used to infer trajectories and dynamic information even without intensive, time-series data by ordering cells according to gene expression similarity. However, while single-cell snapshots sometimes offer valuable insights into dynamic processes, current methods for ordering cells are limited by descriptive notions of "pseudotime" that lack intrinsic physical meaning. Instead of pseudotime, we propose inference of "process time" via a principled modeling approach to formulating trajectories and inferring latent variables corresponding to timing of cells subject to a biophysical process. Our implementation of this approach, called Chronocell, provides a biophysical formulation of trajectories built on cell state transitions. The Chronocell model is identifiable, making parameter inference meaningful. Furthermore, Chronocell can interpolate between trajectory inference, when cell states lie on a continuum, and clustering, when cells cluster into discrete states. By using a variety of datasets ranging from cluster-like to continuous, we show that Chronocell enables us to assess the suitability of datasets and reveals distinct cellular distributions along process time that are consistent with biological process times. We also compare our parameter estimates of degradation rates to those derived from metabolic labeling datasets, thereby showcasing the biophysical utility of Chronocell. Nevertheless, based on performance characterization on simulations, we find that process time inference can be challenging, highlighting the importance of dataset quality and careful model assessment.
Collapse
Affiliation(s)
- Meichen Fang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Gennady Gorin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Lior Pachter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, California, United States of America
| |
Collapse
|
43
|
Medina-Feliciano JG, Valentín-Tirado G, Luna-Martínez K, Beltran-Rivera A, Miranda-Negrón Y, García-Arrarás JE. Single-cell RNA sequencing of the holothurian regenerating intestine reveals the pluripotency of the coelomic epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601561. [PMID: 39005414 PMCID: PMC11244903 DOI: 10.1101/2024.07.01.601561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In holothurians, the regenerative process following evisceration involves the development of a "rudiment" or "anlage" at the injured end of the mesentery. This regenerating anlage plays a pivotal role in the formation of a new intestine. Despite its significance, our understanding of the molecular characteristics inherent to the constituent cells of this structure has remained limited. To address this gap, we employed state-of-the-art scRNA-seq and HCR-FISH analyses to discern the distinct cellular populations associated with the regeneration anlage. Through this approach, we successfully identified thirteen distinct cell clusters. Among these, two clusters exhibit characteristics consistent with putative mesenchymal cells, while another four show features akin to coelomocyte cell populations. The remaining seven cell clusters collectively form a large group encompassing the coelomic epithelium of the regenerating anlage and mesentery. Within this large group of clusters, we recognized previously documented cell populations such as muscle precursors, neuroepithelial cells and actively proliferating cells. Strikingly, our analysis provides data for identifying at least four other cellular populations that we define as the precursor cells of the growing anlage. Consequently, our findings strengthen the hypothesis that the coelomic epithelium of the anlage is a pluripotent tissue that gives rise to diverse cell types of the regenerating intestinal organ. Moreover, our results provide the initial view into the transcriptomic analysis of cell populations responsible for the amazing regenerative capabilities of echinoderms.
Collapse
|
44
|
Lin J, Jiang S, Chen B, Du Y, Qin C, Song Y, Peng Y, Ding M, Wu J, Lin Y, Xu T. Tertiary Lymphoid Structures are Linked to Enhanced Antitumor Immunity and Better Prognosis in Muscle-Invasive Bladder Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2410998. [PMID: 39739621 DOI: 10.1002/advs.202410998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/02/2024] [Indexed: 01/02/2025]
Abstract
The prognosis for muscle-invasive bladder cancer (MIBC) remains poor, and reliable prognostic markers have yet to be identified. Tertiary lymphoid structures (TLS) have been associated with favorable outcomes in certain cancers. However, the relationship between TLS and MIBC remains unclear. A multi-omics approach is utilized, leveraging single-cell RNA sequencing, spatial transcriptomics, bulk RNA sequencing, and immunohistochemistry, to investigate the roles of B cells and TLS in MIBC. These findings indicate that elevated levels of B cells and TLS correlate with improved prognoses in patients with MIBC, aligning with the robust antitumor immune responses observed in the TLS region. From a mechanistic perspective, CXCL13 serves as a critical cytokine for TLS formation in MIBC, primarily secreted by clonally expanded CXCL13+ T cells. This cytokine interacts with the CXCR5 receptor on NR4A2+ B cells, promoting TLS development. Plasma cells arising within the TLS microenvironment predominantly produce the IGHG antibody, potentially enhancing the phagocytic capabilities of C1QC+ macrophages. From an application standpoint, a TLS-specific gene signature is developed that effectively predicts outcomes in MIBC and other cancers. This study highlights the prognostic potential of TLS in MIBC and reveals immune mechanisms, offering insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Jiaxing Lin
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
- Center for Quantitative Biology and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Shan Jiang
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
- Center for Quantitative Biology and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Baoqiang Chen
- Center for Quantitative Biology and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yiqing Du
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Caipeng Qin
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Yuxuan Song
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Yun Peng
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Mengting Ding
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Jilin Wu
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Yihan Lin
- Center for Quantitative Biology and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan, 610213, China
| | - Tao Xu
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| |
Collapse
|
45
|
Abdelbasset M, Saron WAA, Ma D, Rathore APS, Kozaki T, Zhong C, Mantri CK, Tan Y, Tung CC, Tey HL, Chu JJH, Chen J, Ng LG, Wang H, Ginhoux F, St John AL. Differential contributions of fetal mononuclear phagocytes to Zika virus neuroinvasion versus neuroprotection during congenital infection. Cell 2024; 187:7511-7532.e20. [PMID: 39532096 DOI: 10.1016/j.cell.2024.10.028] [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: 07/19/2023] [Revised: 08/08/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Fetal immune cell functions during congenital infections are poorly understood. Zika virus (ZIKV) can vertically transmit from mother to fetus, causing nervous system infection and congenital ZIKV syndrome (CZS). We identified differential functional roles for fetal monocyte/macrophage cell types and microglia in ZIKV dissemination versus clearance using mouse models. Trafficking of ZIKV-infected primitive macrophages from the yolk sac allowed initial fetal virus inoculation, while recruited monocytes promoted non-productive neuroinflammation. Conversely, brain-resident differentiated microglia were protective, limiting infection and neuronal death. Single-cell RNA sequencing identified transcriptional profiles linked to the protective versus detrimental contributions of mononuclear phagocyte subsets. In human brain organoids, microglia also promoted neuroprotective transcriptional changes and infection clearance. Thus, microglia are protective before birth, contrasting with the disease-enhancing roles of primitive macrophages and monocytes. Differential modulation of myeloid cell phenotypes by genetically divergent ZIKVs underscores the potential of immune cells to regulate diverse outcomes during fetal infections.
Collapse
Affiliation(s)
- Muhammad Abdelbasset
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wilfried A A Saron
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Dongliang Ma
- Neuroscience & Behavioral Disorders Programme, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Abhay P S Rathore
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Pathology, Duke University Medical Center, Durham, NC 27705, USA
| | - Tatsuya Kozaki
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Chengwei Zhong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Chinmay Kumar Mantri
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Yingrou Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; National Skin Centre, National Healthcare Group, Singapore, Singapore
| | - Chi-Ching Tung
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Hong Liang Tey
- National Skin Centre, National Healthcare Group, Singapore, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Lai Guan Ng
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine affiliated Renji Hospital, Shanghai, China
| | - Hongyan Wang
- Neuroscience & Behavioral Disorders Programme, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France; Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Ashley L St John
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Pathology, Duke University Medical Center, Durham, NC 27705, USA; SingHealth Duke-NUS Global Health Institute, Singapore, Singapore.
| |
Collapse
|
46
|
Chen X, Chen R, Wu Y, Yu A, Wang F, Ying C, Yin Y, Chen X, Ma L, Fu Y. FABP5+ macrophages contribute to lipid metabolism dysregulation in type A aortic dissection. Int Immunopharmacol 2024; 143:113438. [PMID: 39447410 DOI: 10.1016/j.intimp.2024.113438] [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: 08/17/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Type A aortic dissection (TAAD) is an acute onset disease with a high mortality rate. TAAD is caused by a tear in the aortic intima and subsequent blood infiltration. The most prominent characteristics of TAAD are aortic media degeneration and inflammatory cell infiltration, which disturb the structural integrity and function of nonimmune and immune cells in the aortic wall. However, to date, there is no systematic evaluation of the interactions between nonimmune cells and immune cells and their effects on metabolism in the context of aortic dissection. Here, multiomics, including bulk RNA-seq, single-cell RNA-seq, and lipid metabolomics, was applied to elucidate the comprehensive TAAD lipid metabolism landscape. Normally, monocytes in the stress response state secrete a variety of cytokines. Injured fibroblasts lack the ability to degrade lipids, which is suspected to contribute to a high lipid environment. Macrophages differentiate into fatty acid binding protein 5-positive (FABP5+) macrophages under the stimulation of metabolic substrates. Moreover, the upregulation of Fabp5+ macrophages were retrospectively validated in TAAD model mice and TAAD patients. Finally, Fabp5+ macrophages can generate a wide range of proinflammatory cytokines, which possibly contribute to TAAD pathogenesis.
Collapse
Affiliation(s)
- Xin Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Ruoshi Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yuefeng Wu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; The Lab of Biomed-X, Zhejiang University-University of Edinburgh Institute (ZJU-UoE), School of Medicine, Zhejiang University, Haining 310000, China
| | - Anfeng Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Fei Wang
- GeneChem Technology Co. Ltd., Shanghai 201203, China
| | - Chenxi Ying
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yifei Yin
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Xiaofan Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Liang Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.
| | - Yufei Fu
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou 310006, China.
| |
Collapse
|
47
|
Khoury Damaa M, Serizay J, Balagué R, Boudjema AR, Faucourt M, Delgehyr N, Goh KJ, Lu H, Tan EK, James CT, Faucon C, Mitri R, Bracht DC, Bingle CD, Dunn NR, Arnold SJ, Zaragosi LE, Barbry P, Koszul R, Omran H, Gil-Gómez G, Escudier E, Legendre M, Roy S, Spassky N, Meunier A. Cyclin O controls entry into the cell-cycle variant required for multiciliated cell differentiation. Cell Rep 2024:115117. [PMID: 39740663 DOI: 10.1016/j.celrep.2024.115117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Multiciliated cells (MCCs) ensure fluid circulation in various organs. Their differentiation is marked by the amplification of cilia-nucleating centrioles, driven by a genuine cell-cycle variant, which is characterized by wave-like expression of canonical and non-canonical cyclins such as Cyclin O (CCNO). Patients with CCNO mutations exhibit a subtype of primary ciliary dyskinesia called reduced generation of motile cilia (RGMC). Here, we show that Ccno is activated at the crossroads of the onset of MCC differentiation, the entry into the MCC cell-cycle variant, and the activation of the centriole biogenesis program. Its absence blocks the G1/S-like transition of the cell-cycle variant, interrupts the centriologenesis transcription program, and compromises the production of centrioles and cilia in mouse brain and human respiratory MCCs. Altogether, our study identifies CCNO as a core regulator of entry into the MCC cell-cycle variant and the interruption of this variant as one etiology of RGMC.
Collapse
Affiliation(s)
- Michella Khoury Damaa
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Jacques Serizay
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France; Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, France
| | - Rémi Balagué
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Amélie-Rose Boudjema
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Nathalie Delgehyr
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Kim Jee Goh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
| | - Hao Lu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Ee Kim Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
| | - Cameron T James
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, 61 Biopolis Drive, Singapore 138673, Singapore; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2RX, UK; Singapore-MIT Association for Research and Technology, Critical Analytics for Manufacturing Personalised-Medicine (SMART CAMP), 1 CREATE Way, Singapore 138602, Singapore
| | - Catherine Faucon
- Centre Hospitalier Intercommunal de Créteil, Laboratoire de Microscopie Électronique, Service d'Anatomopathologie, 94010 Créteil, France
| | - Rana Mitri
- Centre Hospitalier Intercommunal de Créteil, Laboratoire de Microscopie Électronique, Service d'Anatomopathologie, 94010 Créteil, France
| | - Diana Carolin Bracht
- Department of General Pediatrics, University Hospital Muenster, 48149 Muenster, Germany
| | - Colin D Bingle
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2RX, UK
| | - Norris Ray Dunn
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore; Skin Research Institute of Singapore, 11 Mandalay Road #17-01 Clinical Sciences Building, Singapore 308232, Singapore
| | - Sebastian J Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstrasse 25, 79104 Freiburg, Germany; Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Schänzlestrasse18, 79104 Freiburg, Germany
| | - Laure-Emmanuelle Zaragosi
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
| | - Pascal Barbry
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France; 3IA Côte d'Azur, 06560 Sophia Antipolis, France
| | - Romain Koszul
- Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, France
| | - Heymut Omran
- Department of General Pediatrics, University Hospital Muenster, 48149 Muenster, Germany
| | - Gabriel Gil-Gómez
- Hospital del Mar Research Institute, Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Estelle Escudier
- Sorbonne Université, Inserm, Childhood genetic diseases UMR_S933, Hôpital Armand-Trousseau, 75012 Paris, France; AP-HP, Sorbonne Université, Hôpital Armand-Trousseau, 75012 Paris, France
| | - Marie Legendre
- Sorbonne Université, Inserm, Childhood genetic diseases UMR_S933, Hôpital Armand-Trousseau, 75012 Paris, France; AP-HP, Sorbonne Université, Hôpital Armand-Trousseau, 75012 Paris, France
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, 61 Biopolis Drive, Singapore 138673, Singapore; Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119288, Singapore
| | - Nathalie Spassky
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Alice Meunier
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France.
| |
Collapse
|
48
|
Serizay J, Khoury Damaa M, Boudjema AR, Balagué R, Faucourt M, Delgehyr N, Noûs C, Zaragosi LE, Barbry P, Spassky N, Koszul R, Meunier A. Cyclin switch tailors a cell cycle variant to orchestrate multiciliogenesis. Cell Rep 2024:115103. [PMID: 39740664 DOI: 10.1016/j.celrep.2024.115103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 01/02/2025] Open
Abstract
Meiosis, endoreplication, and asynthetic fissions are variations of the canonical cell cycle where either replication or mitotic divisions are muted. Here, we identify a cell cycle variantconserved across organs and mammals, where both replication and mitosis are muted, and that orchestrates the differentiation of post-mitotic progenitors into multiciliated cells (MCCs). MCC progenitors reactivate most of the cell cycle transcriptional program but replace the temporal expression of cyclins E2 and A2 with non-canonical cyclins O and A1. In addition, the primary APC/C inhibitor Emi1 is silenced. Re-expressing cyclins E2 and A2 and/or Emi1 can induce partial replication or mitosis. This shows that a cell can co-opt the cell cycle genetic program and regulate only certain elements to qualitatively and quantitatively divert CDK activity toward differentiation rather than division. We propose this cell cycle variant to exploit the existence of a cytoplasmic-or centriolar-CDK threshold lower than the S-phase threshold.
Collapse
Affiliation(s)
- Jacques Serizay
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France; Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, France.
| | - Michella Khoury Damaa
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Amélie-Rose Boudjema
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Rémi Balagué
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Nathalie Delgehyr
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Camille Noûs
- Cogitamus Laboratory, PSL University, Paris, France
| | - Laure-Emmanuelle Zaragosi
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
| | - Pascal Barbry
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
| | - Nathalie Spassky
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Romain Koszul
- Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, France
| | - Alice Meunier
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France.
| |
Collapse
|
49
|
Catta-Preta CMC, Ghosh K, Sacks DL, Ferreira TR. Single-cell atlas of Leishmania development in sandflies reveals the heterogeneity of transmitted parasites and their role in infection. Proc Natl Acad Sci U S A 2024; 121:e2406776121. [PMID: 39700146 DOI: 10.1073/pnas.2406776121] [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: 04/05/2024] [Accepted: 11/03/2024] [Indexed: 12/21/2024] Open
Abstract
Sandfly vectors transmit Leishmania through egestion of parasites into the host skin. The transmissible dose is shaped by Leishmania development in the sandfly gut, described as a sequential differentiation of promastigote morphotypes. Apart from isolated mammal-infective metacyclic promastigotes, little is known about the transcriptional programs and molecular markers for other stages coinhabiting the midgut in mature infections and cotransmitted by the sandfly bite. Here, we elucidate the single-cell transcriptomic complexity of Leishmania major colonizing its natural vector Phlebotomus duboscqi at early and late infection, providing markers for three transmissible stages. Contrary to prevailing models, our analyses indicate a nonlinear developmental progression, with bifurcation into either replicating early metacyclics or attached and detached haptomonads. We demonstrate that haptomonads constitute a key component of the transmitted inoculum and, along with nondividing late metacyclics, are infectious to and exacerbate the pathology in mice. Our single-cell analysis and validated markers will facilitate further studies on the Leishmania life cycle and its implications for vector-to-host transmission dynamics.
Collapse
Affiliation(s)
- Carolina M C Catta-Preta
- Intracellular Parasite Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Kashinath Ghosh
- Intracellular Parasite Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - David L Sacks
- Intracellular Parasite Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Tiago R Ferreira
- Intracellular Parasite Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| |
Collapse
|
50
|
Coren L, Zaffryar-Eilot S, Odeh A, Kaganovsky A, Hasson P. Fibroblast diversification is an embryonic process dependent on muscle contraction. Cell Rep 2024; 43:115034. [PMID: 39636726 DOI: 10.1016/j.celrep.2024.115034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Fibroblasts, the most common cell type found in connective tissues, play major roles in development, homeostasis, regeneration, and disease. Although specific fibroblast subpopulations have been associated with different biological processes, the mechanisms and unique activities underlying their diversity have not been thoroughly examined. Here, we set out to dissect the variation in skeletal-muscle-resident fibroblasts (mrFibroblasts) during development. Our results demonstrate that mrFibroblasts diversify following the transition from embryonic to fetal myogenesis prior to birth. We find that mrFibroblasts segregate into two major subpopulations occupying distinct niches, with interstitial fibroblasts residing between the muscle fibers and delineating fibroblasts sheathing the muscle. We further show that these subpopulations entail distinct cellular dynamics and transcriptomes. Notably, we find that mrFibroblast subpopulations exert distinct regulatory roles on myoblast proliferation and differentiation. Finally, we demonstrate that this diversification depends on muscle contraction. Altogether, these findings establish that mrFibroblasts diversify in a spatiotemporal embryonic process into distinct cell types, entailing different characteristics and roles.
Collapse
Affiliation(s)
- Lavi Coren
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Shelly Zaffryar-Eilot
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Anas Odeh
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Anna Kaganovsky
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Peleg Hasson
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|