1
|
Snoeck HW. Direct megakaryopoiesis. Curr Opin Hematol 2025; 32:213-220. [PMID: 40197720 DOI: 10.1097/moh.0000000000000871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
PURPOSE OF REVIEW Megakaryocytes are large, polyploid cells that produce platelets and originate from hematopoietic stem cells (HSCs) in the bone marrow. While in the classical paradigm, megakaryocytes are generated in a stepwise fashion through increasingly committed progenitor stages, studies using in-vivo barcoding, transplantation, and in-vitro culture have suggested that, in addition, a more direct pathway existed. The relevance of this direct pathway and its functional and phenotypic characteristics were unclear, however. RECENT FINDINGS Recent publications using fate-mapping and single-cell transplantation now unequivocally demonstrate the existence of a direct megakaryocyte differentiation pathway, provide molecular characterization, and indicate distinct roles and regulation of both pathways. The direct pathway originates from a separate subset of 'top' HSCs, is enhanced by hematopoietic stress, inflammation and aging, bypasses multipotential progenitors, may be more active in myeloproliferative neoplasms, and generates phenotypically distinct megakaryocyte progenitors and more reactive platelets. SUMMARY Novel insights into the direct megakaryocyte differentiation pathway provide a deeper understanding of HSC biology, hematological recovery after myeloablation, and aging of the hematopoietic system, and suggest that this pathway may contribute to the increase in thrombotic incidents with age and in myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Hans-Willem Snoeck
- Columbia Center for Stem Cell Therapies/Columbia Center for Human Development, Department of Medicine
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons
- Division of Pulmonary Medicine, Allergy and Critical Care, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
2
|
Cao J, Guo Z, Xu X, Li P, Fang Y, Deng S. Advances in CRISPR-Cas9 in lineage tracing of model animals. Animal Model Exp Med 2025. [PMID: 40491322 DOI: 10.1002/ame2.70033] [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: 11/08/2024] [Accepted: 04/28/2025] [Indexed: 06/11/2025] Open
Abstract
Cell lineage tracing is a key technology for describing the developmental history of individual progenitor cells and assembling them to form a lineage development tree. However, traditional methods have limitations of poor stability and insufficient resolution. As an efficient and flexible gene editing tool, CRISPR-Cas9 system has been widely used in biological research. Furthermore, CRISPR-Cas9 gene editing-based tracing methods can introduce fluorescent proteins, reporter genes, or DNA barcodes for high-throughput sequencing, enabling precise lineage analysis, significantly improving precision and resolution, and expanding its application range. In this review, we summarize applications of CRISPR-Cas9 system in cell lineage tracing, with special emphasis on its successful applications in traditional model animals (e.g., zebrafish and mice), large animal models (pigs), and human cells or organoids. We also discussed its potential prospects and challenges in xenotransplantation and regenerative medicine.
Collapse
Affiliation(s)
- Jingchao Cao
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zihang Guo
- National Center of Technology Innovation for Animal Model, National Human Diseases Animal Model Resource Center, National Health Commission of China (NHC) Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Xueling Xu
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Pan Li
- Xianghu Laboratory, Hangzhou, China
| | - Yi Fang
- Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Shoulong Deng
- National Center of Technology Innovation for Animal Model, National Human Diseases Animal Model Resource Center, National Health Commission of China (NHC) Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Herstine JA, Mensh J, Coffman E, George SM, Herman K, Martin JB, Zatari A, Chandler HL, Kozmik Z, Drysdale TA, Bridgewater D, Plageman TF. Shroom3 facilitates optic fissure closure via tissue alignment and reestablishment of apical-basal polarity during epithelial fusion. Dev Biol 2025; 522:91-105. [PMID: 40113025 DOI: 10.1016/j.ydbio.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/24/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Optic cup morphogenesis is a complex process involving cellular behaviors such as epithelial folding, cell shape changes, proliferation, and tissue fusion. Disruptions to these processes can lead to an ocular coloboma, a congenital defect where the optic fissure fails to close. This study investigates the role of Shroom3, a protein implicated in epithelial morphogenesis, in mouse embryos during optic cup development. It was observed that Shroom3 is apically localized in the neural retina and retinal pigmented epithelium, and its deficiency leads to a both a conventional coloboma phenotype characterized by a gap in pigmented tissue as well as a unique type of coloboma where an ectopic ventral fold of neural tissue is present. Increased apical areas of both neural retina and retinal pigmented epithelial cells are present in the absence of Shroom3 leading to a greater apical surface area and disruption of optic fissure alignment. Neural retina specific gene ablation revealed that Shroom3 function in the RPE is likely sufficient to facilitate tissue alignment and permit fusion. However, the fusion process is ultimately disturbed due to a failure of the neural tissue to reestablish apical-basal polarity. Furthermore, it is demonstrated that Shroom3 deficiency also affects other epithelial fusion events in the embryo that rely on polarity reestablishment, such as lens vesicle separation, eyelid formation, and secondary palate closure. These findings highlight the importance of Shroom3 during optic cup morphogenesis, aid our understanding of optic fissure closure and coloboma formation, and implicates a role for Shroom3 in regulating apical-basal polarity.
Collapse
Affiliation(s)
| | - Jordyn Mensh
- College of Optometry, The Ohio State University, Columbus, OH, USA
| | - Electra Coffman
- College of Optometry, The Ohio State University, Columbus, OH, USA
| | | | - Kenneth Herman
- College of Optometry, The Ohio State University, Columbus, OH, USA
| | - Jessica B Martin
- College of Optometry, The Ohio State University, Columbus, OH, USA
| | - Ali Zatari
- College of Optometry, The Ohio State University, Columbus, OH, USA
| | | | - Zbynek Kozmik
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Thomas A Drysdale
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Darren Bridgewater
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
4
|
Lee SM, Cichanski SR, Pintozzi NG, Kaufmann M, Jones G, Meyer MB. Kidney deletions of Cyp27b1 fail to reduce serum 1,25(OH) 2D 3. J Steroid Biochem Mol Biol 2025; 250:106734. [PMID: 40096920 DOI: 10.1016/j.jsbmb.2025.106734] [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: 12/27/2024] [Revised: 02/17/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Vitamin D metabolism is controlled through the kidney mitochondrial P450 enzymes 1α-hydroxylase (CYP27B1) and 24-hydroxylase (CYP24A1) that activate and degrade the endocrine vitamin D hormone (1,25(OH)2D3), respectively. We recently demonstrated that extrarenal cells can make 1,25(OH)2D3 with adequate vitamin D supplementation by targeted mass spectrometry imaging in our Cyp27b1 kidney enhancer deletion mouse model that lacks circulating 1,25(OH)2D3 (M1/M21-DIKO mouse). Based on these observations, we selectively deleted Cyp27b1 (Cyp27b1fl/fl) from the mouse kidney using the Six2- and Pax8-cre drivers that target tubule and nephron development to see if we could recapitulate the remarkable phenotype of the M1/M21-DIKO mice. While Six2-cre/Cyp27b1fl/fl mice had a mild phenotype, Pax8-cre/Cyp27b1fl/fl mice had a marked elevation of parathyroid hormone and a reduction in bone mineral density. The vitamin D metabolic profile in the Pax8-cre/Cyp27b1fl/fl clearly indicated a dysfunction in the CYP24A1 enzyme with reductions in 24,25(OH)2D3 and 25(OH)D3-26,23-lactone with an accompanying elevation of 25(OH)D3. However, despite these compensatory reductions in CYP24A1 derived metabolites and apparent deletion of Cyp27b1 in the kidney, the 1,25(OH)2D3 levels were not changed from wildtype in either mouse. Like 24,25(OH)2D3, the 1,24,25(OH)3D3 levels were also reduced. These data highlight the robust homeostatic mechanisms to salvage 1,25(OH)2D3, point towards potential compensatory mechanisms of 1,25(OH)2D3 production from non-kidney tissues, and reinforce the utility of the M1/M21-DIKO model as a non-global deletion of Cyp27b1 with reductions in serum 1,25(OH)2D3 to be used to understand the complexity of vitamin D metabolism in health and inflammatory disease.
Collapse
Affiliation(s)
- Seong Min Lee
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Shannon R Cichanski
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Nicolas G Pintozzi
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Martin Kaufmann
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L3N6, Canada; Department of Surgery, Queen's University, Kingston, Ontario K7L3N6, Canada
| | - Glenville Jones
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L3N6, Canada
| | - Mark B Meyer
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
5
|
Bisia AM, Xypolita ME, Bikoff EK, Robertson EJ, Costello I. Eomesodermin in conjunction with the BAF complex promotes expansion and invasion of the trophectoderm lineage. Nat Commun 2025; 16:5079. [PMID: 40450029 DOI: 10.1038/s41467-025-60417-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 05/23/2025] [Indexed: 06/03/2025] Open
Abstract
The T-box transcription factor (TF) Eomesodermin/Tbr2 (Eomes) is essential for maintenance of the trophectoderm (TE) lineage, but the molecular mechanisms underlying this critical role remain obscure. Here, we show in trophoblast stem cells (TSCs) that Eomes partners with several TE-specific TFs as well as chromatin remodellers, including Brg1 and other subunits of the BAF complex. Degron-mediated Eomes protein depletion results in genome-wide loss of chromatin accessibility at TSC-specific loci. These overlap with a subset of sites that lose accessibility following Brg1 inhibition, suggesting that Eomes acts as a "doorstop" controlling TSC chromatin accessibility. Eomes depletion also causes transcriptional misregulation of TSC maintenance and early differentiation markers. An additional subset of Eomes-dependent genes encode intercellular/matricellular interaction and cytoskeletal components, likely explaining the implantation defects of Eomes-null embryos. Thus, Eomes promotes TE lineage maintenance by sustaining trophectoderm-specific chromatin accessibility, while promoting the gene regulatory networks that modulate expansion and cell behaviour during implantation.
Collapse
Affiliation(s)
| | | | | | | | - Ita Costello
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
6
|
Marchand T, Akinnola KE, Takeishi S, Maryanovich M, Pinho S, Saint-Vanne J, Birbrair A, Lamy T, Tarte K, Frenette P, Gritsman K. Periosteal skeletal stem cells can migrate into the bone marrow and support hematopoiesis after injury. eLife 2025; 13:RP101714. [PMID: 40401637 PMCID: PMC12097789 DOI: 10.7554/elife.101714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
Skeletal stem cells (SSCs) have been isolated from various tissues, including periosteum and bone marrow, where they exhibit key functions in bone biology and hematopoiesis, respectively. The role of periosteal SSCs (P-SSCs) in bone regeneration and healing has been extensively studied, but their ability to contribute to the bone marrow stroma is still under debate. In the present study, we characterized a mouse whole bone transplantation model that mimics the initial bone marrow necrosis and fatty infiltration seen after injury. Using this model and a lineage tracing approach, we observed the migration of P-SSCs into the bone marrow after transplantation. Once in the bone marrow, P-SSCs are phenotypically and functionally reprogrammed into bone marrow mesenchymal stem cells (BM-MSCs) that express high levels of hematopoietic stem cell niche factors such as Cxcl12 and Kitl. In addition, using ex vivo and in vivo approaches, we found that P-SSCs are more resistant to acute stress than BM-MSCs. These results highlight the plasticity of P-SSCs and their potential role in bone marrow regeneration after bone marrow injury.
Collapse
Affiliation(s)
- Tony Marchand
- Service d’hématologie Clinique, Centre Hospitalier Universitaire de RennesRennesFrance
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
| | - Kemi E Akinnola
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
| | - Shoichiro Takeishi
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
| | - Maria Maryanovich
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
| | - Sandra Pinho
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Medical Oncology, Albert Einstein College of MedicineBronxUnited States
- Department of Pharmacology & Regenerative Medicine, University of Illinois at ChicagoChicagoUnited States
| | - Julien Saint-Vanne
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
| | - Alexander Birbrair
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Dermatology, University of Wisconsin-MadisonMadisonUnited States
| | - Thierry Lamy
- Service d’hématologie Clinique, Centre Hospitalier Universitaire de RennesRennesFrance
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
| | - Karin Tarte
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
- Laboratoire Suivi Immunologique des Thérapeutiques Innovantes, Centre Hospitalier Universitaire de RennesRennesFrance
| | - Paul Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Medical Oncology, Albert Einstein College of MedicineBronxUnited States
| | - Kira Gritsman
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Medical Oncology, Albert Einstein College of MedicineBronxUnited States
| |
Collapse
|
7
|
Dang Y, Lattner J, Lahola-Chomiak AA, Afonso DA, Ulbricht E, Taubenberger A, Rulands S, Tabler JM. Self-propagating wave drives morphogenesis of skull bones in vivo. Nat Commun 2025; 16:4330. [PMID: 40346043 PMCID: PMC12064835 DOI: 10.1038/s41467-025-59164-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/13/2025] [Indexed: 05/11/2025] Open
Abstract
Cellular motion is a key feature of tissue morphogenesis and is often driven by migration. However, migration need not explain cell motion in contexts where there is little free space or no obvious substrate, such as those found during organogenesis of mesenchymal organs including the embryonic skull. Through ex vivo imaging, biophysical modeling, and perturbation experiments, we find that mechanical feedback between cell fate and stiffness drives bone expansion and controls bone size in vivo. This mechanical feedback system is sufficient to propagate a wave of differentiation that establishes a collagen gradient which we find sufficient to describe patterns of osteoblast motion. Our work provides a mechanism for coordinated motion that may not rely upon cell migration but on emergent properties of the mesenchymal collective. Identification of such alternative mechanisms of mechanochemical coupling between differentiation and morphogenesis will help in understanding how directed cellular motility arises in complex environments with inhomogeneous material properties.
Collapse
Affiliation(s)
- Yiteng Dang
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
- Center for Systems Biology, Dresden, Germany
| | - Johanna Lattner
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Diana Alves Afonso
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | | | | | - Steffen Rulands
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
- Center for Systems Biology, Dresden, Germany
- Arnold-Sommerfeld-Center for Theoretical Physics, Ludwig-Maximilians-Universität München, München, Germany
| | - Jacqueline M Tabler
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
8
|
Recka N, Simons A, Cornell RA, Van Otterloo E. Epidermal loss of PRMT5 leads to the emergence of an atypical basal keratinocyte-like cell population and defective skin stratification. J Invest Dermatol 2025:S0022-202X(25)00449-X. [PMID: 40339790 DOI: 10.1016/j.jid.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 05/10/2025]
Abstract
During skin development, ectoderm-derived cells undergo precisely coordinated proliferation, differentiation, and adhesion to yield stratified epidermis. Disruptions in these processes can result in congenital anomalies including ectodermal dysplasia and harlequin ichthyosis. Protein Arginine Methyl Transferase 5 (PRMT5)-an enzyme responsible for methylating arginine residues in histones and other proteins-maintains progenitor status in germ and limb bud cells. Similarly, in vitro evidence suggests that PRMT5 prevents differentiation of basal keratinocytes, leading us to hypothesize that PRMT5 preserves the stem-cell phenotype of keratinocytes in vivo. To test this possibility, we generated conditional knockout (cKO) mice lacking Prmt5 in early ectoderm (E7.5), impacting the entire epidermis. Prmt5 cKOs exhibited gross skin defects, compromised skin barrier function, and reduced postnatal viability. Histological analyses revealed significant defects in epidermal stratification, without alterations in apoptosis or proliferation. Single-cell RNA and ATAC-seq analysis identified an atypical population of basal keratinocyte-like cells in Prmt5 cKOs, that exhibited a senescence-like program, characterized by increased Cdkn1a (p21), elevated senescence-associated secretory phenotype (SASP) molecules (Igfbp2), and decreased developmental transcription factor (Trp63) expression. Our findings suggest that PRMT5 prevents basal keratinocyte senescence by repressing Cdkn1a, shedding light on the epigenetic regulation of basal keratinocyte maintenance and senescence in congenital skin disorders.
Collapse
Affiliation(s)
- Nicole Recka
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA; Iowa Institute of Oral Health Research, College of Dentistry & Dental Clinics, University of Iowa, Iowa City, IA, 52242, USA; Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52242, USA
| | - Andrean Simons
- Department of Radiation Oncology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Robert A Cornell
- Department of Oral Health Sciences, University of Washington, School of Dentistry, Seattle, WA, 98195, USA
| | - Eric Van Otterloo
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA; Iowa Institute of Oral Health Research, College of Dentistry & Dental Clinics, University of Iowa, Iowa City, IA, 52242, USA; Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52242, USA; Department of Periodontics, College of Dentistry & Dental Clinics, University of Iowa, Iowa City, IA, 52242, USA; Craniofacial Anomalies Research Center, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
9
|
Cleary SJ, Qiu L, Seo Y, Baluk P, Liu D, Serwas NK, Taylor CA, Zhang D, Cyster JG, McDonald DM, Krummel MF, Looney MR. Intravital imaging of pulmonary lymphatics in inflammation and metastatic cancer. J Exp Med 2025; 222:e20241359. [PMID: 39969509 PMCID: PMC11837973 DOI: 10.1084/jem.20241359] [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: 08/03/2024] [Revised: 12/11/2024] [Accepted: 01/24/2025] [Indexed: 02/20/2025] Open
Abstract
Intravital microscopy has enabled the study of immune dynamics in the pulmonary microvasculature, but many key events remain unseen because they occur in deeper lung regions. We therefore developed a technique for stabilized intravital imaging of bronchovascular cuffs and collecting lymphatics surrounding pulmonary veins in mice. Intravital imaging of pulmonary lymphatics revealed ventilation dependence of steady-state lung lymph flow and ventilation-independent lymph flow during inflammation. We imaged the rapid exodus of migratory dendritic cells through lung lymphatics following inflammation and measured effects of pharmacologic and genetic interventions targeting chemokine signaling. Intravital imaging also captured lymphatic immune surveillance of lung-metastatic cancers and lymphatic metastasis of cancer cells. To our knowledge, this is the first imaging of lymph flow and leukocyte migration through intact pulmonary lymphatics. This approach will enable studies of protective and maladaptive processes unfolding within the lungs and in other previously inaccessible locations.
Collapse
Affiliation(s)
- Simon J. Cleary
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Institute of Pharmaceutical Science, King’s College London, London, UK
| | - Longhui Qiu
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Yurim Seo
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Peter Baluk
- Department of Anatomy, Cardiovascular Research Institute, and Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
| | - Dan Liu
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA
- Westlake Laboratory of Life Sciences and Biomedicine, School of Medicine, Westlake University, Hangzhou, China
| | | | - Catherine A. Taylor
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Dongliang Zhang
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Jason G. Cyster
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA
- Bakar ImmunoX Initiative, UCSF, San Francisco, CA, USA
| | - Donald M. McDonald
- Department of Anatomy, Cardiovascular Research Institute, and Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, USA
| | - Matthew F. Krummel
- Department of Pathology, UCSF, San Francisco, CA, USA
- Bakar ImmunoX Initiative, UCSF, San Francisco, CA, USA
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Bakar ImmunoX Initiative, UCSF, San Francisco, CA, USA
| |
Collapse
|
10
|
Banjac I, Maimets M, Tsang IHC, Dioli M, Hansen SL, Krizic K, Bressan RB, Lövkvist C, Jensen KB. Fate mapping in mouse demonstrates early secretory differentiation directly from Lgr5+ intestinal stem cells. Dev Cell 2025; 60:1281-1289.e6. [PMID: 39793582 DOI: 10.1016/j.devcel.2024.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 08/10/2024] [Accepted: 12/11/2024] [Indexed: 01/13/2025]
Abstract
The intestinal epithelium has a remarkably high turnover in homeostasis. It remains unresolved how this is orchestrated at the cellular level and how the behavior of stem and progenitor cells ensures tissue maintenance. To address this, we combined quantitative fate mapping in three complementary mouse models with mathematical modeling and single-cell RNA sequencing. Our integrated approach generated a spatially and temporally defined model of crypt maintenance based on two cycling populations: stem cells at the crypt-bottom and transit-amplifying (TA) cells above them. Subsequently, we validated the predictions from the mathematical model, demonstrating that fate decisions between the secretory and absorptive lineages are made within the stem cell compartment, whereas TA cell divisions contribute specifically to the absorptive lineage. These quantitative insights provide further direct evidence for crypt-bottom stem cells as the dominant driver of the intestinal epithelium replenishment.
Collapse
Affiliation(s)
- Isidora Banjac
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Martti Maimets
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Ingrid H C Tsang
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Marius Dioli
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Stine Lind Hansen
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Kata Krizic
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Raul Bardini Bressan
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Cecilia Lövkvist
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.
| | - Kim B Jensen
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.
| |
Collapse
|
11
|
Dong K, Bai Z, He X, Zhang L, Hu G, Yao Y, Cai CL, Zhou J. Generation of a novel constitutive smooth muscle cell-specific Myh11-driven Cre mouse model. J Mol Cell Cardiol 2025; 202:144-152. [PMID: 40122158 DOI: 10.1016/j.yjmcc.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Dysfunction in either embryonic or postnatal smooth muscle cells (SMCs) significantly contributes to the progression of various cardiovascular and visceral diseases. Therefore, elucidating the molecular mechanisms governing SMC development and homeostasis is crucial. MYH11 is the most reliable lineage gene for SMCs and has been utilized to develop tamoxifen-inducible Cre driver lines for achieving SMC-specific gene manipulation by crossing with mice carrying the loxP-flanked gene, particularly in adult mice. For studies involving SMCs during embryogenesis, the commonly used constitutive Cre driver is controlled by the Tagln (also known as SM22α) promoter. However, this Cre driver exhibits activity in multiple non-SMC populations, including cardiomyocytes and skeletal muscle precursors, introducing confounding effects. Additionally, most existing SMC-specific Cre drivers are generated using a transgenic approach, raising concerns about random site integration and variable gene copy numbers. To address these limitations, we report a novel Cre mouse model generated by knock-in (KI) of a nuclear-localized Cre recombinase into the Myh11 gene locus using homologous recombination. We confirmed that the Cre activity precisely recapitulates endogenous Myh11 expression by crossing with Rosa26 mTmG or tdTomato reporter mice. Moreover, Myh11-driven Cre can efficiently delete the floxed allele of the transcription factor Tead1 specifically in SMCs. The Tead1 SMC-specific knockout mice did not exhibit an overt phenotype, thereby circumventing the embryonic lethal phenotype mediated by Tagln-driven Cre, as we previously reported. These findings establish this novel Cre driver line as a robust tool for tracing the Myh11-positive SMC lineage and manipulating gene function specifically in SMCs during embryonic development in both male and female mice.
Collapse
Affiliation(s)
- Kunzhe Dong
- Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA; Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Zhixia Bai
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Department of Anesthesiology and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Xiangqin He
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Lu Zhang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Guoqing Hu
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yali Yao
- Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Chen-Leng Cai
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China.
| | - Jiliang Zhou
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
12
|
Merle C, Rodrigues C, Pourkhalili Langeroudi A, Journot R, Rost F, Dang Y, Rulands S, Fre S. Transcriptional landscapes underlying Notch-induced lineage conversion and plasticity of mammary basal cells. EMBO J 2025; 44:2827-2855. [PMID: 40186028 DOI: 10.1038/s44318-025-00424-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 04/07/2025] Open
Abstract
The mammary epithelium derives from multipotent mammary stem cells (MaSCs) that engage into differentiation during embryonic development. However, adult MaSCs maintain the ability to reactivate multipotency in non-physiological contexts. We previously reported that Notch1 activation in committed basal cells triggers a basal-to-luminal cell fate switch in the mouse mammary gland. Here, we report conservation of this mechanism and found that in addition to the mammary gland, constitutive Notch1 signaling induces a basal-to-luminal cell fate switch in adult cells of the lacrimal gland, the salivary gland, and the prostate. Since the lineage transition is progressive in time, we performed single-cell transcriptomic analysis on index-sorted mammary cells at different stages of lineage conversion, generating a temporal map of changes in cell identity. Combining single-cell analyses with organoid assays, we demonstrate that cell proliferation is indispensable for this lineage conversion. We also reveal the individual transcriptional landscapes underlying the cellular plasticity switching of committed mammary cells in vivo with spatial and temporal resolution. Given the roles of Notch signaling in cancer, these results may help to better understand the mechanisms that drive cellular transformation.
Collapse
Affiliation(s)
- Candice Merle
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Calvin Rodrigues
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Atefeh Pourkhalili Langeroudi
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Robin Journot
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France
| | - Fabian Rost
- Max-Planck-Institute for the Physics of Complex Systems, Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | - Yiteng Dang
- Max-Planck-Institute for the Physics of Complex Systems, Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
- Center for Systems Biology, Dresden, Germany
| | - Steffen Rulands
- Ludwig-Maximilians-Universität München, Arnold-Sommerfeld-Center for Theoretical Physics, München, Germany
| | - Silvia Fre
- Institut Curie, Laboratory of Genetics and Developmental Biology, INSERM U934, CNRS UMR3215, PSL University, Sorbonne University, Paris, France.
| |
Collapse
|
13
|
Bluemn T, Kessler JR, Kim AJ, Drnevich J, Lincoln J. Murine Aortic Valve Cell Heterogeneity at Birth. Arterioscler Thromb Vasc Biol 2025; 45:743-753. [PMID: 40079140 DOI: 10.1161/atvbaha.124.322280] [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: 12/05/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Heart valve function requires a highly organized ECM (extracellular matrix) network that provides the necessary biomechanical properties needed to withstand pressure changes during each cardiac cycle. Lay down of the valve ECM begins during embryogenesis and continues throughout postnatal stages when it is remodeled into stratified layers and arranged according to blood flow. Alterations in this process can lead to dysfunction and, if left untreated, heart failure. Despite this, the mechanisms that establish structure-function relationships of the valve, particularly during postnatal maturation, are poorly understood. METHODS To address this, single-cell transcriptomics was performed on murine aortic valve structures at postnatal day 1. RESULTS Overall, 18 clusters of 7 diverse cell populations were identified, including a novel valve endothelial cell subpopulation unique to postnatal day 1 and 3 previously unappreciated valve interstitial cell subpopulations defined as primitive, remodeling, and bioactive. Additional lineage tracing of the primitive valve interstitial cell subpopulation in mice uncovered a temporal and spatial trajectory throughout postnatal maturation. CONCLUSIONS In summary, this work highlights the heterogeneity of cell types within the aortic valve structure at birth that contribute to establishing and maintaining structure and function throughout life.
Collapse
Affiliation(s)
- Theresa Bluemn
- Department of Pediatrics, Division of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee (T.B., J.R.K., A.J.K., J.L.)
- Herma Heart Institute, Children's Wisconsin, Milwaukee (T.B., J.R.K., A.J.K., J.L.)
| | - Julie R Kessler
- Department of Pediatrics, Division of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee (T.B., J.R.K., A.J.K., J.L.)
- Herma Heart Institute, Children's Wisconsin, Milwaukee (T.B., J.R.K., A.J.K., J.L.)
| | - Andrew J Kim
- Department of Pediatrics, Division of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee (T.B., J.R.K., A.J.K., J.L.)
- Herma Heart Institute, Children's Wisconsin, Milwaukee (T.B., J.R.K., A.J.K., J.L.)
| | - Jenny Drnevich
- High Performance Computing in Biology, Roy J. Carver Biotechnology Center, University of Illinois Urbana-Champaign (J.D.)
| | - Joy Lincoln
- Department of Pediatrics, Division of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee (T.B., J.R.K., A.J.K., J.L.)
- Herma Heart Institute, Children's Wisconsin, Milwaukee (T.B., J.R.K., A.J.K., J.L.)
| |
Collapse
|
14
|
Song L, Wang L, He Z, Cui X, Peng C, Xu J, Yong Z, Liu Y, Fei JF. Improving Spatial Transcriptomics with Membrane-Based Boundary Definition and Enhanced Single-Cell Resolution. SMALL METHODS 2025; 9:e2401056. [PMID: 39871658 DOI: 10.1002/smtd.202401056] [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: 07/10/2024] [Revised: 01/03/2025] [Indexed: 01/29/2025]
Abstract
Accurately defining cell boundaries for spatial transcriptomics is technically challenging. The current major approaches are nuclear staining or mathematical inference, which either exclude the cytoplasm or determine a hypothetical boundary. Here, a new method is introduced for defining cell boundaries: labeling cell membranes using genetically coded fluorescent proteins, which allows precise indexing of sequencing spots and transcripts within cells on sections. Use of this membrane-based method greatly increases the number of genes captured in cells compared to the number captured using nucleus-based methods; the numbers of genes are increased by 67% and 119% in mouse and axolotl livers, respectively. The obtained expression profiles are more consistent with single-cell RNA-seq data, demonstrating more rational clustering and apparent cell type-specific markers. Furthermore, improved single-cell resolution is achieved to better identify rare cell types and elaborate spatial domains in the axolotl brain and intestine. In addition to regular cells, accurate recognition of multinucleated cells and cells lacking nuclei in the mouse liver is achieved, demonstrating its ability to analyze complex tissues and organs, which is not achievable using previous methods. This study provides a powerful tool for improving spatial transcriptomics that has broad potential for its applications in the biological and medical sciences.
Collapse
Affiliation(s)
- Li Song
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Liqun Wang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Zitian He
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Xiao Cui
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Cheng Peng
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Jie Xu
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Zhouying Yong
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Yanmei Liu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 510631, China
| | - Ji-Feng Fei
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
15
|
Tuwatnawanit T, Wessman W, Belisova D, Sumbalova Koledova Z, Tucker A, Anthwal N. FSP1/S100A4-Expressing Stem/Progenitor Cells Are Essential for Temporomandibular Joint Growth and Homeostasis. J Dent Res 2025; 104:551-560. [PMID: 39953712 PMCID: PMC12000630 DOI: 10.1177/00220345251313795] [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: 02/17/2025] Open
Abstract
The temporomandibular joint (TMJ) is one of the most used joints in the body. Defects and wear in the cartilage of the joint, condyle, and fibrocartilage disc lie at the heart of many common TMJ disorders. During postnatal development, the condyle acts as a growth center for the mandible, with cells moving as a conveyor belt away from the top of the condyle as they differentiate. The superficial layers of the condyle have been proposed to contain stem/progenitor populations to allow growth and maintain homeostasis. Here we have focused on the role of fibroblast-specific protein 1 (FSP1; also known as S100a4) as a key fibroblast stem/progenitor marker for the condyle. Lineage tracing with FSP1-Cre;R26RmTmG mice revealed that FSP1-expressing cells were restricted to the superficial fibroblast zone, giving rise to all layers of the condyle over time. The FSP1-expressing cells overlapped with other putative stem cell markers of the condyle, such as Gli1 and scleraxis. BrdU pulse chase experiments highlighted that a subset of FSP1 fibrocartilage was label retaining, suggesting that FSP1 labels a novel stem/progenitor cell population in the condyle. Destruction of FSP1-expressing cells by conditional diphtheria toxin activity in FSP1-Cre;R26RDTA mice resulted in severe TMJ osteoarthritis with loss of the cartilage structure. Lgr5-expressing cells in the superficial layer of the condyle have been shown to create a Wnt inhibitory niche. FSP1 expression postnatally was associated with a reduction in canonical Wnt activity in the condyle. Importantly, constitutive activation of Wnt/β catenin in FSP1-expressing cells led to a downregulation of FSP1 and progressive postnatal loss of TMJ condylar hyaline cartilage due to loss of the superficial stem/progenitor cells. These data demonstrate a novel role for FSP1-expressing cells in the superficial zone in growth and maintenance of the TMJ condylar cartilage and highlight the importance of regulating Wnt activity in this population.
Collapse
Affiliation(s)
- T. Tuwatnawanit
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, UK
- Department of Conservative Dentistry and Prosthodontics, Faculty of Dentistry, Srinakharinwirot University, Wattana, Bangkok, Thailand
| | - W. Wessman
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - D. Belisova
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Z. Sumbalova Koledova
- Laboratory of Tissue Morphogenesis and Cancer, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - A.S. Tucker
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, UK
| | - N. Anthwal
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, UK
| |
Collapse
|
16
|
Abou Nader N, Jakuc N, Meinsohn MC, Charrier L, Banville L, Brind’Amour J, Paquet M, St-Jean G, Boerboom D, Mao J, Pépin D, Breault DT, Zamberlam G, Boyer A. Hippo Signaling Is Essential for the Maintenance of Zona Glomerulosa Cell Fate in the Murine Adrenal Cortex. Endocrinology 2025; 166:bqaf077. [PMID: 40233139 PMCID: PMC12041920 DOI: 10.1210/endocr/bqaf077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/14/2025] [Accepted: 04/14/2025] [Indexed: 04/17/2025]
Abstract
Cells of the zona glomerulosa (zG), the outermost zone of the adrenal cortex, secrete aldosterone and transdifferentiate into glucocorticoid-producing cells of the zona fasciculata (zF) during adrenal homeostasis. However, our understanding of the signaling pathways mediating zG cell maintenance or their transdifferentiation into zF cells is incomplete. Hippo is a major pathway that regulates cell proliferation/differentiation during embryogenesis and postnatal tissue homeostasis. Hypothesizing that Hippo signaling could be involved in zG cell maintenance or transdifferentiation, we generated a mouse model in which the two main kinases of the Hippo signaling cascade large tumor suppressor homolog kinases 1/2 (Lats1 and Lats2) are specifically inactivated in zG cells. Here we show that loss of function of Lats1 and Lats2 impairs zG steroidogenesis and leads to zG cell transdifferentiation into cells sharing characteristics with chondroblasts/osteoblasts rather than zF cells. Furthermore, we demonstrate that this phenotype can be rescued by the concomitant inactivation of the transcriptional coactivators Yes-associated protein (Yap) and transcriptional coactivator with PDZ-binding motif (Taz) with Lats1 and Lats2. Finally, we show that expression of a constitutively active form of YAP (YAP5SA) in zG cells does not alter their fate as severely as the loss of Lats1 and Lats2 but leads to adrenal hyperplasia. Together, these findings highlight the critical role of Hippo signaling in maintaining zG cell fate and function and provide key insights into broader mechanisms underlying cellular differentiation.
Collapse
Affiliation(s)
- Nour Abou Nader
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Natalia Jakuc
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | | | - Laureline Charrier
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Laurence Banville
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Pathologie et de Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Julie Brind’Amour
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Marilène Paquet
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Pathologie et de Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Guillaume St-Jean
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Pathologie et de Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David T Breault
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Gustavo Zamberlam
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Alexandre Boyer
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| |
Collapse
|
17
|
Wen H, Chandrasekaran P, Jin A, Pankin J, Lu M, Liberti DC, Zepp JA, Jain R, Morrisey EE, Michki SN, Frank DB. A spatiotemporal cell atlas of cardiopulmonary progenitor cell allocation during development. Cell Rep 2025; 44:115513. [PMID: 40178979 PMCID: PMC12103214 DOI: 10.1016/j.celrep.2025.115513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 01/10/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
The heart and lung co-orchestrate their development during organogenesis. The mesoderm surrounding both the developing heart and anterior foregut endoderm provides instructive cues guiding cardiopulmonary development. Additionally, it serves as a source of cardiopulmonary progenitor cells (CPPs) expressing Wnt2 that give rise to both cardiac and lung mesodermal cell lineages. Despite the mesoderm's critical importance to both heart and lung development, mechanisms guiding CPP specification are unclear. To address this, we lineage traced Wnt2+ CPPs at E8.5 and performed single-cell RNA sequencing on collected progeny across the developmental lifespan. Using computational analyses, we created a CPP-derived cell atlas that revealed a previously underappreciated spectrum of CPP-derived cell lineages, including all lung mesodermal lineages, ventricular cardiomyocytes, and epicardial and pericardial cells. By integrating spatial mapping with computational cell trajectory analysis and transcriptional profiling, we have provided a potential molecular and cellular roadmap for cardiopulmonary development.
Collapse
Affiliation(s)
- Hongbo Wen
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Prashant Chandrasekaran
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Annabelle Jin
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Josh Pankin
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - MinQi Lu
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Derek C Liberti
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Jarod A Zepp
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, University of Pennsylvania, CHOP, Penn-CHOP Lung Biology Institute, Philadelphia, PA 19104, USA
| | - Rajan Jain
- Department of Medicine, Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Department of Medicine, Department of Cell and Developmental Biology, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sylvia N Michki
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA; Department of Pediatrics, Division of Pulmonary and Sleep Medicine, University of Pennsylvania, CHOP, Penn-CHOP Lung Biology Institute, Philadelphia, PA 19104, USA.
| | - David B Frank
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Tian X, Yang G, Zheng H, Pi Y, Cao Z, Duan P, Chen Z, Yuan G. The Mdm2-p53 axis links cementocyte survival to cellular cementum volume. J Bone Miner Res 2025; 40:548-560. [PMID: 39903703 DOI: 10.1093/jbmr/zjaf025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/26/2024] [Accepted: 01/31/2025] [Indexed: 02/06/2025]
Abstract
Cementocytes are terminally differentiated cells embedded in cellular cementum, an important hard tissue covering the apical regions of tooth roots. However, the roles of cementocytes in cellular cementum remain enigmatic. Here, we show that Murine Double Minute 2 (Mdm2), an E3 ubiquitin ligase that plays vital roles in regulating cell proliferation, apoptosis, and differentiation to influence tissue or organ development, is highly expressed in the cementocytes of mice. To investigate the role of cementocyte-expressed Mdm2, Dmp1-Cre;Mdm2flox/flox (Mdm2 cKO)mice were obtained to inactivate Mdm2 in cementocytes. The results showed that Mdm2 was successfully ablated and Mdm2 cKO mice display increased cementocyte apoptosis and reduced cellular cementum volume. p53, the canonical substrate of Mdm2, was accumulated and hyperactivated in the cementocytes of Mdm2 cKO mice and in cultured IDG-CM6 cells (a cementocyte cell line) treated with Nutlin3a, an inhibitor of Mdm2. Further experiments showed that inactivation of 1 allele of p53 significantly rescued the increased cementocyte apoptosis and the decreased cellular cementum volume in Mdm2 cKO mice. Therefore, p53 is targeted by Mdm2 for degradation and mediates the role of Mdm2 in cementocyte survival and cellular cementum volume. Notably, Mdm2 cKO mice exhibited decreased differentiation of cementoblasts (the cell type primarily responsible for cementum deposition) and reduced rate of cellular cementum deposition. Meanwhile, OCCM-30 cells (a cementoblast cell line) showed diminished migration, proliferation, differentiation, and mineralization ability after culture with conditioned medium (CM) from Nutlin3a-pretreated IDG-CM6 cells. Intriguingly, Mdm2 cKO mice displayed significantly increased osteoclast formation and cementum resorption. Meanwhile, in vitro experiments verified that CM from Nutlin3a-pretreated IDG-CM6 cells induced osteoclast differentiation of bone marrow macrophages. Collectively, these results demonstrate that Mdm2-mediated degradation of p53 promotes cementocyte survival, and that cementocytes affect the cell behaviors of cementoblasts and osteoclasts through a paracrine mode to modulate cellular cementum volume.
Collapse
Affiliation(s)
- Xue Tian
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Guobin Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Huiwen Zheng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Yixing Pi
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Zhengguo Cao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Peipei Duan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhi Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Guohua Yuan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| |
Collapse
|
19
|
Wu Z, Wang Y, Chen WW, Sun H, Chen X, Li X, Wang Z, Liang W, Wang SY, Luan X, Li Y, Huang S, Liang Y, Zhang J, Chen ZF, Wang G, Gao Y, Liu Y, Wang J, Liu Z, Shi P, Liu C, Lv L, Hou A, Wu C, Yao C, Hong Z, Dai J, Lu Z, Pan F, Chen X, Kettenmann H, Amit I, Speakman JR, Chen Y, Ginhoux F, Cui R, Huang T, Li H. Peripheral nervous system microglia-like cells regulate neuronal soma size throughout evolution. Cell 2025; 188:2159-2174.e15. [PMID: 40199320 DOI: 10.1016/j.cell.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 11/18/2024] [Accepted: 02/12/2025] [Indexed: 04/10/2025]
Abstract
Microglia, essential in the central nervous system (CNS), were historically considered absent from the peripheral nervous system (PNS). Here, we show a PNS-resident macrophage population that shares transcriptomic and epigenetic profiles as well as an ontogenetic trajectory with CNS microglia. This population (termed PNS microglia-like cells) enwraps the neuronal soma inside the satellite glial cell envelope, preferentially associates with larger neurons during PNS development, and is required for neuronal functions by regulating soma enlargement and axon growth. A phylogenetic survey of 24 vertebrates revealed an early origin of PNS microglia-like cells, whose presence is correlated with neuronal soma size (and body size) rather than evolutionary distance. Consistent with their requirement for soma enlargement, PNS microglia-like cells are maintained in vertebrates with large peripheral neuronal soma but absent when neurons evolve to have smaller soma. Our study thus reveals a PNS counterpart of CNS microglia that regulates neuronal soma size during both evolution and ontogeny.
Collapse
Affiliation(s)
- Zhisheng Wu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Department of Immunology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; School of Chemistry and Chemical Engineering, Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yiheng Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wei-Wei Chen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hua Sun
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; School of Life Sciences, Henan University, Henan, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Xiaobo Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zeshuai Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weizheng Liang
- Hebei Provincial Key Laboratory of Systems Biology and Gene Regulation, Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Shuang-Yin Wang
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Xuemei Luan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yijiang Li
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Shangjin Huang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuteng Liang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jiaqi Zhang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhou-Feng Chen
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, and Shenzhen Medical Academy of Research and Translation, Shenzhen, China
| | - Guanlin Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China; Shanghai Qi Zhi Institute, Shanghai, China
| | - Yun Gao
- State Key Laboratory of Genetic Resources and Evolution, and Southwest Research Centre of Porcine Molecular Breeding and Translational Medicine in China, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yanan Liu
- State Key Laboratory of Genetic Resources and Evolution, and Southwest Research Centre of Porcine Molecular Breeding and Translational Medicine in China, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jun Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Liu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Peng Shi
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Cirong Liu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Longbao Lv
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Anli Hou
- Shenzhen Guangming District People's Hospital, Shenzhen, China
| | - Chenglin Wu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chen Yao
- The First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, Shenzhen, China
| | - Zexuan Hong
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ji Dai
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhonghua Lu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fan Pan
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | | | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Yun Chen
- Department of Immunology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; School of Chemistry and Chemical Engineering, Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| | - Florent Ginhoux
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Rongfeng Cui
- School of Ecology & State Key Laboratory of Biocontrol, Sun Yat-sen University, Shenzhen, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, China
| | - Tianwen Huang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hanjie Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen University of Advanced Technology, Shenzhen, China.
| |
Collapse
|
20
|
Wiesner E, Binz-Lotter J, Tröder SE, Unnersjö-Jess D, Rutkowski N, Zevnik B, Schermer B, Benzing T, Wedlich-Söldner R, Hackl MJ. A red fluorescent lifeact marker to study actin morphology in podocytes. Sci Rep 2025; 15:12386. [PMID: 40216917 PMCID: PMC11992033 DOI: 10.1038/s41598-025-96822-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: 09/17/2024] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
F-actin is a major component of the cellular cytoskeleton, responsible for maintaining cell shape, enabling movement and facilitating intracellular transport. In the kidney, glomerular podocytes are highly dependent on their actin cytoskeleton shaping their unique foot processes. Hereditary mutations in actin-binding proteins cause focal segmental glomerulosclerosis, while other organs remain largely unaffected. So far, actin visualization in podocytes has been limited to electron microscopy or indirect immunofluorescent labeling of actin-binding proteins. However, the short F-actin-binding peptide Lifeact enables researchers to study actin dynamics in vitro and in vivo with minimal interference with actin metabolism. Here we introduce a new mouse model with conditional expression of a Lifeact.mScarlet-I fusion protein providing red labeling of actin. Cre recombinase-mediated activity allows cell-specific and mosaic expression in podocytes, enabling selective labeling of individual cells to contrast with non-expressing neighboring cells. Transgenic mice are born healthy and young animals display no kidney-related phenotype. By intravital imaging and super-resolution microscopy, we show subcellular localization of actin to the foot processes in a resolution previously only obtainable by electron microscopy. Our novel mouse line provides the opportunity to study the actin cytoskeleton in podocytes and other cell types by intravital imaging and other conventional light microscopy techniques.
Collapse
Affiliation(s)
- Eva Wiesner
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Julia Binz-Lotter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Simon E Tröder
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- In vivo Research Facility, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - David Unnersjö-Jess
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Nelli Rutkowski
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Branko Zevnik
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- In vivo Research Facility, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Roland Wedlich-Söldner
- Institute of Cell Dynamics and Imaging and Cells in Motion Interfaculty Centre, University of Münster, Münster, Germany
| | - Matthias J Hackl
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
21
|
Chen X, Shao J, Brandenburger I, Qian W, Hahnefeld L, Bonnavion R, Cho H, Wang S, Hidalgo J, Wettschureck N, Geisslinger G, Gurke R, Wang Z, Offermanns S. FFAR4-mediated IL-6 release from islet macrophages promotes insulin secretion and is compromised in type-2 diabetes. Nat Commun 2025; 16:3422. [PMID: 40210633 PMCID: PMC11986018 DOI: 10.1038/s41467-025-58706-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 03/20/2025] [Indexed: 04/12/2025] Open
Abstract
The function of islet macrophages is poorly understood. They promote glucose-stimulated insulin secretion (GSIS) in lean mice, however, the underlying mechanism has remained unclear. We show that activation of the free fatty acid receptor FFAR4 on islet macrophages leads to interleukin-6 (IL-6) release and that IL-6 promotes β-cell function. This mechanism is required for GSIS in lean male mice, but does not function anymore in islets from people with obesity and obese type 2 diabetic male mice. In islets from obese mice, FFAR4 downstream signaling in macrophages is strongly reduced, resulting in impaired FFAR4-mediated IL-6 release. However, IL-6 treatment can still improve GSIS in islets from people with obesity and obese type 2 diabetic mice. These data show that a defect in FFAR4-mediated macrophage activation contributes to reduced GSIS in type 2 diabetes and suggest that reactivating islet macrophage FFAR4 and promoting or mimicking IL-6 release from islet macrophages improves GSIS in type 2 diabetes.
Collapse
Affiliation(s)
- Xinyi Chen
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Jingchen Shao
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Isabell Brandenburger
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Weikun Qian
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Pancreas Center of Xi'an Jiaotong University, Xi'an, China
| | - Lisa Hahnefeld
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP) and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
- Goethe University Frankfurt, Institute of Clinical Pharmacology, Faculty of Medicine, Frankfurt, Germany
| | - Rémy Bonnavion
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Haaglim Cho
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - ShengPeng Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University. Xi'an, Shaanxi, China
| | - Juan Hidalgo
- Department of Cellular Biology, Physiology, and Immunology, Autonomous University of Barcelona, Barcelona, Spain
| | - Nina Wettschureck
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
- Center for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Excellence Cluster Cardiopulmonary Institute (CPI), Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt/Rhine-Main, Bad Nauheim, Germany
| | - Gerd Geisslinger
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP) and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
- Goethe University Frankfurt, Institute of Clinical Pharmacology, Faculty of Medicine, Frankfurt, Germany
| | - Robert Gurke
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP) and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
- Goethe University Frankfurt, Institute of Clinical Pharmacology, Faculty of Medicine, Frankfurt, Germany
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Pancreas Center of Xi'an Jiaotong University, Xi'an, China
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany.
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University. Xi'an, Shaanxi, China.
- Center for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany.
- Excellence Cluster Cardiopulmonary Institute (CPI), Bad Nauheim, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt/Rhine-Main, Bad Nauheim, Germany.
| |
Collapse
|
22
|
Reese CF, Gooz M, Hajdu Z, Hoffman S. CD45+/ Col I+ Fibrocytes: Major source of collagen in the fibrotic lung, but not in passaged fibroblast cultures. Matrix Biol 2025; 136:87-101. [PMID: 39828137 DOI: 10.1016/j.matbio.2025.01.005] [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: 07/09/2024] [Revised: 01/06/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
The role of cells of the hematopoietic lineage in fibrosis is controversial. Here we evaluate the contribution of Col I+/CD45+ cells (fibrocytes) to lung fibrosis. Systemic bleomycin treatment was used to induce fibrosis in a bone marrow transplant and two transgenic mouse models. Lung cells from these mice were analyzed by flow cytometry, both immediately upon release from the tissue or following growth on tissue-culture plastic. Fibrotic and control human lung tissue were also used. Fibroblasts and fibrocytes derived from a transgenic mouse model were compared in terms of their morphology, growth, and adhesion to fibronectin. Single cell RNAseq was performed with the analysis focusing on CD45-/Col I+ "fibroblasts" and CD45+/Col I+ "fibrocytes" in control and fibrotic mouse lung tissue. Finally, we inhibited fibrosis in mice using a novel, water-soluble version of caveolin scaffolding domain (CSD) called WCSD. In both mouse and human lung tissue, we observed by flow cytometry a large increase in fibrocyte number and Col I expression associated with fibrosis. In contrast, fibroblast number was not significantly increased. A large increase (>50-fold) in fibrocyte number associated with fibrosis was also observed by single cell RNAseq. In this case, fibroblasts increased 5-fold. Single cell RNAseq also revealed that myofibroblast markers in fibrotic tissue are associated with a cluster containing a similar number of fibrocytes and fibroblasts, not with a resident fibroblast cluster. Some investigators claim that fibrocytes are not present among primary fibroblasts. However, we found that fibrocytes were the predominant cell type present in these cultures prior to passage. Fewer fibrocytes were present after one passage, and almost none after two passages. Our experiments suggest that fibrocytes are crowded out of cultures during passage because fibroblasts have a larger footprint than fibrocytes, even though fibrocytes bind more efficiently to fibronectin. Finally, we observed by flow cytometry that in mice treated with bleomycin and WCSD compared to bleomycin alone, there was a large decrease in the number of fibrocytes present but not in the number of fibroblasts. In summary, fibrocytes are a major collagen-producing cell type that is increased in number in association with fibrosis as well as a major source of myofibroblasts. The common observation that collagen-producing spindle-shaped cells associated with fibrosis are CD45- may be an artifact of passage in cell culture.
Collapse
Affiliation(s)
- Charles F Reese
- Division of Rheumatology/Department of Medicine, Medical University of South Carolina, Charleston 29425, SC, USA
| | - Monika Gooz
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston 29425, SC, USA
| | - Zoltan Hajdu
- Department of Anatomical Sciences, Edward Via College of Osteopathic Medicine, 350 Howard Street, Spartanburg 29303, SC, USA
| | - Stanley Hoffman
- Division of Rheumatology/Department of Medicine, Medical University of South Carolina, Charleston 29425, SC, USA.
| |
Collapse
|
23
|
Han D, Xu W, Jeong HW, Park H, Weyer K, Tsytsyura Y, Stehling M, Wu G, Lan G, Kim KP, Renner H, Han DW, Chen Y, Gerovska D, Araúzo-Bravo MJ, Klingauf J, Schwamborn JC, Adams RH, Liu P, Schöler HR. Multipotent neural stem cells originating from neuroepithelium exist outside the mouse central nervous system. Nat Cell Biol 2025; 27:605-618. [PMID: 40211073 PMCID: PMC11991921 DOI: 10.1038/s41556-025-01641-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/19/2025] [Indexed: 04/12/2025]
Abstract
Conventional understanding dictates that mammalian neural stem cells (NSCs) exist only in the central nervous system. Here, we report that peripheral NSCs (pNSCs) exist outside the central nervous system and can be isolated from mouse embryonic limb, postnatal lung, tail, dorsal root ganglia and adult lung tissues. Derived pNSCs are distinct from neural crest stem cells, express multiple NSC-specific markers and exhibit cell morphology, self-renewing and differentiation capacity, genome-wide transcriptional profile and epigenetic features similar to control brain NSCs. pNSCs are composed of Sox1+ cells originating from neuroepithelial cells. pNSCs in situ have similar molecular features to NSCs in the brain. Furthermore, many pNSCs that migrate out of the neural tube can differentiate into mature neurons and limited glial cells during embryonic and postnatal development. Our discovery of pNSCs provides previously unidentified insight into the mammalian nervous system development and presents an alternative potential strategy for neural regenerative therapy.
Collapse
Affiliation(s)
- Dong Han
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany.
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Wan Xu
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hyun-Woo Jeong
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Hongryeol Park
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Kathrin Weyer
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Yaroslav Tsytsyura
- Department of Cellular Biophysics, Institute for Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Martin Stehling
- Flow Cytometry Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Guangming Wu
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Division of Basic Research, Guangzhou National Laboratory, Guangzhou, China
| | - Guocheng Lan
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kee-Pyo Kim
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Henrik Renner
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | | | - Yicong Chen
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Daniela Gerovska
- Group of Computational Biology and Systems Biomedicine, Biogipuzkoa Health Research Institute, San Sebastian, Spain
| | - Marcos J Araúzo-Bravo
- Group of Computational Biology and Systems Biomedicine, Biogipuzkoa Health Research Institute, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jürgen Klingauf
- Department of Cellular Biophysics, Institute for Medical Physics and Biophysics, University of Münster, Münster, Germany
- IZKF Münster and Cluster of Excellence EXC 1003, Cells in Motion (CiM), Münster, Germany
| | - Jens Christian Schwamborn
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- University of Münster, Medical Faculty, Münster, Germany
| | - Pentao Liu
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Center for Translational Stem Cell Biology, Hong Kong, China.
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany.
| |
Collapse
|
24
|
Ali A, Yun S. Multifaceted Role of Notch Signaling in Vascular Health and Diseases. Biomedicines 2025; 13:837. [PMID: 40299408 PMCID: PMC12024539 DOI: 10.3390/biomedicines13040837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Notch signaling is evolutionarily conserved from Drosophila to mammals and it functions as an essential modulator of vascular growth and development by directing endothelial cell specification, proliferation, migration, arteriovenous differentiation, inflammation, and apoptosis. The interplay between Notch and other signaling pathways plays a homeostatic role by modulating multiple vascular functions, including permeability regulation, angiogenesis, and vascular remodeling. This review explores current knowledge on Notch signaling in vascular development, homeostasis, and disease. It also discusses recent developments in understanding how endothelial Notch signaling affects vascular inflammation via cytokines or aberrant shear stress in endothelial cells while addressing the reciprocal relationship between Notch signaling and inflammation.
Collapse
Affiliation(s)
| | - Sanguk Yun
- Department of Biotechnology, Inje University, Gimhae 50834, Republic of Korea;
| |
Collapse
|
25
|
Iliopoulou L, Tzaferis C, Prados A, Roumelioti F, Koliaraki V, Kollias G. Different fibroblast subtypes propel spatially defined ileal inflammation through TNFR1 signalling in murine ileitis. Nat Commun 2025; 16:3023. [PMID: 40155385 PMCID: PMC11953319 DOI: 10.1038/s41467-025-57570-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 02/25/2025] [Indexed: 04/01/2025] Open
Abstract
Crohn's disease (CD) is a persistent inflammatory disorder primarily affecting the terminal ileum. The TnfΔΑRE mice, which spontaneously develop CD-like ileitis due to TNF overexpression, represent a faithful model of the human disease. Here, via single-cell RNA sequencing in TnfΔΑRE mice, we show that murine TNF-dependent ileitis is characterized by cell expansion in tertiary lymphoid organs (TLO), T cell effector reprogramming, and accumulation of activated macrophages in the submucosal granulomas. Within the stromal cell compartment, fibroblast subsets (telocytes, trophocytes, PdgfraloCd81- cells) are less abundant while lymphatic endothelial cells (LEC) and fibroblastic reticular cells (FRC) show relative expansion compared to the wild type. All three fibroblast subsets show strong pro-inflammatory signature. TNFR1 loss or gain of function experiments in specific fibroblast subsets suggest that the TnfΔΑRE-induced ileitis is initiated in the lamina propria via TNF pathway activation in villus-associated fibroblasts (telocytes and PdgfraloCd81- cells), which are responsible for the organization of TLOs. Trophocytes drive disease progression in the submucosal layer, accompanied by the excessive formation of granulomas. These findings provide evidence for spatial regulation of inflammation by fibroblast subsets and underscore the pivotal role of fibroblasts in the inception and advancement of ileitis.
Collapse
Affiliation(s)
- Lida Iliopoulou
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Christos Tzaferis
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Alejandro Prados
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Fani Roumelioti
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Vasiliki Koliaraki
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - George Kollias
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
26
|
Suthakaran S, Homami S, Chavez D, Tang S, Moore SKL, Sussman C, Zhang J, Britto CJ, Prince A, May AJ, Kathiriya JJ, Hook JL. CFTR function in alveolar type 1 cells drives lung liquid secretion and host defense. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645303. [PMID: 40196491 PMCID: PMC11974899 DOI: 10.1101/2025.03.25.645303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Loss of the liquid layer that lines the lung's air-facing surface underpins mechanisms of major lung diseases, but the development of therapies that restore liquid secretion is hampered by an incomplete understanding of the cell types that drive it. Here, we show CFTR function in alveolar type 1 (AT1) cells - a cell type that comprises 95% of the lung surface but is presumed to be unimportant in CFTR-related diseases - is critical to lung liquid secretion and the secretion-mediated clearance of particles and S. aureus from lung alveoli. Our findings reveal essential roles for AT1 cells in lung homeostasis and defense, and they call for a reevaluation of the role of AT1 cells in CFTR-related diseases. We suggest AT1 cells be considered key targets of secretion-restoring therapies.
Collapse
|
27
|
Guo Z, Yao J, Zheng X, Cao J, Lv X, Gao Z, Guo S, Li H, Guan D, Li L, Qin D, Li D, Wang X, Tan M, Zhang J, Zhang Y, Wang B, Bu W, Li J, Zhao X, Meng F, Feng Y, Li L, Du J, Fan Y. Cavity oscillation drives pattern formation in early mammalian embryos. Cell Rep 2025; 44:115342. [PMID: 39985766 DOI: 10.1016/j.celrep.2025.115342] [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: 04/23/2024] [Revised: 10/02/2024] [Accepted: 01/31/2025] [Indexed: 02/24/2025] Open
Abstract
During the second cell fate in mouse embryos, the inner cell mass (ICM) segregates into the spatially distinct epiblast (EPI) and primitive endoderm (PrE) layers. The mechanism driving this pattern formation, however, remains unresolved. Here, we report that, concomitant with the segregation process of EPI/PrE precursors starting from mid-blastocyst, the blastocyst cavity begins to oscillate cyclically with rapid contraction yet slow expansion, triggering a phase transition in the ICM to a fluid-like state. This asymmetric oscillation of the blastocyst cavity facilitates EPI/PrE segregation by enhancing cell-cell contact fluctuations within the ICM and initiating convergent cell flows, which induce movement of these two cell types in opposite directions, wherein PrE precursors move toward the ICM-lumen interface, whereas EPI precursors move toward the trophectoderm. Last, we found that both PDGFRα expression and YAP nuclear accumulation in PrE precursors increase in response to blastocyst cavity oscillation. This study reveals the foundational role of physical oscillation in driving embryonic pattern formation during early mammalian embryonic development.
Collapse
Affiliation(s)
- Zheng Guo
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Jie Yao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xu Zheng
- State Key Laboratory of Nonlinear Mechanics, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Jialing Cao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xinxin Lv
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Zheng Gao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Shuyu Guo
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Hangyu Li
- State Key Laboratory of Nonlinear Mechanics, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Dongshi Guan
- State Key Laboratory of Nonlinear Mechanics, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Long Li
- State Key Laboratory of Nonlinear Mechanics, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Dandan Qin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dong Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaoxiao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Min Tan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Jing Zhang
- Laboratory Animal Research Center, Tsinghua University, Beijing 100084, China
| | - Yanli Zhang
- Imaging Core Facility, Technology Center for Protein Science, Tsinghua University, Beijing 100084, China
| | - Bo Wang
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xi'ning 810008, China
| | - Wanjuan Bu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Jianwen Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xinbin Zhao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Fanzhe Meng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yue Feng
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jing Du
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| |
Collapse
|
28
|
Chin HS, Cheng J, Hsu SH, Lum GG, Zaldivia MT, Nelameham S, Guo F, Mallavarapu K, Jackling FC, Yang J, Tan JSL, Sampath P, Barker N, Smyth GK, Lindeman GJ, Strasser A, Visvader JE, Chen Y, Chen T, Fu NY. MCL‑1 safeguards activated hair follicle stem cells to enable adult hair regeneration. Nat Commun 2025; 16:2829. [PMID: 40121237 PMCID: PMC11929845 DOI: 10.1038/s41467-025-58150-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
Hair follicles cycle through expansion, regression and quiescence. To investigate the role of MCL‑1, a BCL‑2 family protein with anti‑apoptotic and apoptosis‑unrelated functions, we delete Mcl‑1 within the skin epithelium using constitutive and inducible systems. Constitutive Mcl‑1 deletion does not impair hair follicle organogenesis but leads to gradual hair loss and elimination of hair follicle stem cells. Acute Mcl‑1 deletion rapidly depletes activated hair follicle stem cells and completely blocks depilation‑induced hair regeneration in adult mice, while quiescent hair follicle stem cells remain unaffected. Single‑cell RNA‑seq profiling reveals the engagement of P53 and DNA mismatch repair signaling in hair follicle stem cells upon depilation‑induced activation. Trp53 deletion rescues hair regeneration defects caused by acute Mcl‑1 deletion, highlighting a critical interplay between P53 and MCL‑1 in balancing proliferation and death. The ERBB pathway plays a central role in sustaining the survival of adult activated hair follicle stem cells by promoting MCL‑1 protein expression. Remarkably, the loss of a single Bak allele, a pro‑apoptotic Bcl‑2 effector gene, rescues Mcl‑1 deletion‑induced defects in both hair follicles and mammary glands. These findings demonstrate the pivotal role of MCL‑1 in inhibiting proliferation stress‑induced apoptosis when quiescent stem cells activate to fuel tissue regeneration.
Collapse
Affiliation(s)
- Hui San Chin
- Cancer and Stem Cell Biology Program, Duke‑NUS Medical School, Singapore, Singapore.
| | - Jinming Cheng
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Shih Han Hsu
- Cancer and Stem Cell Biology Program, Duke‑NUS Medical School, Singapore, Singapore
| | - Guo Guang Lum
- Cancer and Stem Cell Biology Program, Duke‑NUS Medical School, Singapore, Singapore
| | - Maria Tk Zaldivia
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Sarmilla Nelameham
- Cancer and Stem Cell Biology Program, Duke‑NUS Medical School, Singapore, Singapore
| | - Fusheng Guo
- Cancer and Stem Cell Biology Program, Duke‑NUS Medical School, Singapore, Singapore
| | | | - Felicity C Jackling
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Jicheng Yang
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Jonathan S L Tan
- A*STAR Skin Research Labs (A*SRL), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Prabha Sampath
- Cancer and Stem Cell Biology Program, Duke‑NUS Medical School, Singapore, Singapore
- A*STAR Skin Research Labs (A*SRL), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Nick Barker
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, Australia
| | - Geoffrey J Lindeman
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
- The Royal Melbourne Hospital, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Andreas Strasser
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Jane E Visvader
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Yunshun Chen
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Ting Chen
- National Institute of Biological Sciences, Beijing, China
| | - Nai Yang Fu
- Cancer and Stem Cell Biology Program, Duke‑NUS Medical School, Singapore, Singapore.
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
29
|
Zhu S, Huo S, He W, Huang C, Zhang J, Jiang X, Qian Y, Chen C, Dai ZM, Yang X, Qiu M, Li T, Zhu XJ. Fine-tuning of Wnt signaling by RNA surveillance factor Smg5 in the mouse craniofacial development. iScience 2025; 28:111972. [PMID: 40071146 PMCID: PMC11894330 DOI: 10.1016/j.isci.2025.111972] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/21/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
The specific roles of nonsense-mediated mRNA decay (NMD), a translation-dependent RNA quality control mechanism that degrades mRNAs containing premature termination codons (PTCs), in mammalian craniofacial development have remained unclear. Here, we show that knockout of the essential NMD factor Smg5 in mouse craniofacial neural crest cells leads to hypoplastic mandibles, subsequently inducing tongue mispositioning and cleft palate formation. Furthermore, Smg5 loss triggers massive cell apoptosis and disrupts cell differentiation, accompanied by widespread alterations in alternative splicing and a surge in PTC-containing mRNA levels. Notably, the abnormal upregulation of a PTC-containing Porcn transcript leads to reduced Porcn protein and impaired Wnt5a/JNK signaling, a crucial pathway for craniofacial morphogenesis. Finally, death of Smg5-deficient craniofacial neural crest cells can be ameliorated by Wnt5a in craniofacial neural crest (CNC) in vitro explants. Taken together, our findings demonstrate that Smg5-mediated NMD regulates mammalian craniofacial development by fine-tuning Wnt signaling through post-transcriptional regulation of Porcn.
Collapse
Affiliation(s)
- Shicheng Zhu
- College of Life and Environmental Sciences, Zhejiang Key Laboratory of Organ Development and Regeneration, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Suman Huo
- College of Life and Environmental Sciences, Zhejiang Key Laboratory of Organ Development and Regeneration, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Weiran He
- College of Life and Environmental Sciences, Zhejiang Key Laboratory of Organ Development and Regeneration, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Caiyan Huang
- College of Life and Environmental Sciences, Zhejiang Key Laboratory of Organ Development and Regeneration, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jiannan Zhang
- College of Life and Environmental Sciences, Zhejiang Key Laboratory of Organ Development and Regeneration, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xiaoning Jiang
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yeqing Qian
- Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Chengyan Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 250100, China
| | - Zhong-Min Dai
- College of Life and Environmental Sciences, Zhejiang Key Laboratory of Organ Development and Regeneration, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xueqin Yang
- College of Life and Environmental Sciences, Zhejiang Key Laboratory of Organ Development and Regeneration, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Mengsheng Qiu
- College of Life and Environmental Sciences, Zhejiang Key Laboratory of Organ Development and Regeneration, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Tangliang Li
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 250100, China
| | - Xiao-Jing Zhu
- College of Life and Environmental Sciences, Zhejiang Key Laboratory of Organ Development and Regeneration, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
30
|
Deng Y, He Y, Xu J, He H, Zhang M, Li G. Cardiac Fibroblasts regulate myocardium and coronary vasculature development via the collagen signaling pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.11.612512. [PMID: 39314489 PMCID: PMC11418987 DOI: 10.1101/2024.09.11.612512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The fibroblast (FB), cardiomyocyte (CM), and vascular endothelial cell (Vas_EC) are the three major cell types in the heart, yet their relationships during development are largely unexplored. To address this gap, we employed RNA staining of the FB marker gene Col1a1 together with the CM marker gene Actn2 and the Vas_EC marker gene Cdh5 at different stages. This approach enabled us to discern the anatomical pattern of cardiac FBs and identify approximately one EC and four CMs directly interacting with each FB. Molecularly, through the analysis of single-cell mRNA sequencing (scRNA-seq) data, we unveiled collagen as the top signaling molecule derived from FBs influencing CM and Vas_EC development. Subsequently, we used a Pdgfra-CreER controlled diphtheria toxin A (DTA) system to ablate the FBs at different stages. We found that the ablation of FBs disrupted myocardium and vasculature development and led to embryonic heart defects. Using scRNA-seq, we further profiled the ablated hearts and identified molecular defects in their ventricular CMs and Vas_ECs compared to control hearts. Moreover, we identified a reduction of collagen in the ablated hearts and predicted collagen as the major signaling pathway regulating the differentially expressed genes in the ablated ventricular CMs. Finally, we performed both short-term and long-term fibroblast ablation at the neonatal stage. We found that short-term ablation caused a reduction in collagen and Vas_EC density, while long-term ablation may induce compensatory collagen expression without causing heart function reduction. In summary, our study has identified the function of fibroblasts in regulating myocardium and vasculature development and implicated an important role for the collagen pathway in this process.
Collapse
Affiliation(s)
- Yiting Deng
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Yuanhang He
- Tsinghua University, Tsinghua medicine, School of Medicine, Beijing, China
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Juan Xu
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Haoting He
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Manling Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Guang Li
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| |
Collapse
|
31
|
Hsu AY, Huang Q, Pi X, Fu J, Raghunathan K, Ghimire L, Balasubramanian A, Xie X, Yu H, Loison F, Haridas V, Zha J, Liu F, Park SY, Bagale K, Ren Q, Fan Y, Zheng Y, Cancelas JA, Chai L, Stowell SR, Chen K, Xu R, Wang X, Xu Y, Zhang L, Cheng T, Ma F, Thiagarajah JR, Wu H, Feng S, Luo HR. Neutrophil-derived vesicles control complement activation to facilitate inflammation resolution. Cell 2025; 188:1623-1641.e26. [PMID: 39938514 PMCID: PMC11934499 DOI: 10.1016/j.cell.2025.01.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/23/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025]
Abstract
Although subsets with immunosuppressive properties exist, neutrophils are typically known for their pro-inflammatory role and pathogen clearance capabilities. Here, we reveal that neutrophils can paradoxically aid in resolving inflammation by actively producing anti-inflammatory extracellular vesicles. These large aging-neutrophil-derived vesicles (LAND-Vs) do not fit into classical vesicle categorizations due to their specific size, structure, or biogenesis pathway. They are protected from efferocytotic clearance by phagocytes due to surface "do not eat me" signals and accumulate in the resolution phase of inflammation. CD55 on LAND-Vs exerts a robust, sustained anti-inflammatory effect by inhibiting complement 3 convertase, thereby reducing neutrophil recruitment and tissue damage. CD55+ LAND-Vs originate in ordered lipid raft domains, where CD55 accumulates asymmetrically during neutrophil aging, and are subsequently formed through RhoA-dependent budding. Collectively, LAND-V emerges as a pivotal physiological immunomodulator and showcases functions that transcend the limited lifespan of neutrophils, offering a therapeutic target for inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Alan Y Hsu
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Qingxiang Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Xiong Pi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 20115, USA
| | - Jianing Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 20115, USA
| | - Krishnan Raghunathan
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA 20115, USA
| | - Laxman Ghimire
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Arumugam Balasubramanian
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Xuemei Xie
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Hongbo Yu
- Department of Pathology and Laboratory Medicine, VA Boston Healthcare System, West Roxbury, Boston, MA 02132, USA
| | - Fabien Loison
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Viraga Haridas
- Flow and Imaging Cytometry Resources, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jiali Zha
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Fei Liu
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Shin-Young Park
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Kamal Bagale
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yuping Fan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yi Zheng
- Experimental Hematology and Cancer Biology Research, Cincinnati Children's Hospital Medical Center, Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Jose A Cancelas
- Experimental Hematology and Cancer Biology Research, Cincinnati Children's Hospital Medical Center, Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Li Chai
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Sean R Stowell
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Kanchao Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Rong Xu
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Xiaoxue Wang
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yuanfu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Lianghui Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Fengxia Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Jay R Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA 20115, USA; Congenital Enteropathy Program, Boston Children's Hospital, PediCODE Consortium, Harvard Digestive Disease Center, Boston, MA, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 20115, USA
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China.
| | - Hongbo R Luo
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Anbarci DN, McKey J, Levic DS, Bagnat M, Capel B. Rediscovering the rete ovarii, a secreting auxiliary structure to the ovary. eLife 2025; 13:RP96662. [PMID: 40105200 PMCID: PMC11922502 DOI: 10.7554/elife.96662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
The rete ovarii (RO) is an appendage of the ovary that has been given little attention. Although the RO appears in drawings of the ovary in early versions of Gray's Anatomy, it disappeared from recent textbooks, and is often dismissed as a functionless vestige in the adult ovary. Using PAX8 immunostaining and confocal microscopy, we characterized the fetal development of the RO in the context of the mouse ovary. The RO consists of three distinct regions that persist in adult life, the intraovarian rete (IOR), the extraovarian rete (EOR), and the connecting rete (CR). While the cells of the IOR appear to form solid cords within the ovary, the EOR rapidly develops into a convoluted tubular epithelium ending in a distal dilated tip. Cells of the EOR are ciliated and exhibit cellular trafficking capabilities. The CR, connecting the EOR to the IOR, gradually acquires tubular epithelial characteristics by birth. Using microinjections into the distal dilated tip of the EOR, we found that luminal contents flow toward the ovary. Mass spectrometry revealed that the EOR lumen contains secreted proteins potentially important for ovarian function. We show that the cells of the EOR are closely associated with vasculature and macrophages, and are contacted by neuronal projections, consistent with a role as a sensory appendage of the ovary. The direct proximity of the RO to the ovary and its integration with the extraovarian landscape suggest that it plays an important role in ovary development and homeostasis.
Collapse
Affiliation(s)
- Dilara N Anbarci
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| | - Jennifer McKey
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Daniel S Levic
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| | - Michel Bagnat
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| |
Collapse
|
33
|
Moore Zajic EL, Zhao R, McKinney MC, Yi K, Wood C, Trainor PA. Cell extrusion drives neural crest cell delamination. Proc Natl Acad Sci U S A 2025; 122:e2416566122. [PMID: 40063802 PMCID: PMC11929498 DOI: 10.1073/pnas.2416566122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/17/2025] [Indexed: 03/15/2025] Open
Abstract
Neural crest cells (NCC) comprise a heterogeneous population of cells with variable potency that contribute to nearly every tissue and organ throughout the body. Considered unique to vertebrates, NCC are transiently generated within the dorsolateral region of the neural plate or neural tube during neurulation. Their delamination and migration are crucial for embryo development as NCC differentiation is influenced by their final resting locations. Previous work in avian and aquatic species revealed that NCC delaminate via an epithelial-mesenchymal transition (EMT), which transforms these progenitor cells from static polarized epithelial cells into migratory mesenchymal cells with fluid front and back polarity. However, the cellular and molecular mechanisms facilitating NCC delamination in mammals are poorly understood. Through time-lapse imaging of NCC delamination in mouse embryos, we identified a subset of cells that exit the neuroepithelium as isolated round cells, which then halt for a short period prior to acquiring the mesenchymal migratory morphology classically associated with delaminating NCC. High-magnification imaging and protein localization analyses of the cytoskeleton, together with measurements of pressure and tension of delaminating NCC and neighboring neuroepithelial cells, revealed that round NCC are extruded from the neuroepithelium prior to completion of EMT. Furthermore, cranial NCC are extruded through activation of the mechanosensitive ion channel, PIEZO1. Our results support a model in which cell density, pressure, and tension in the neuroepithelium result in activation of the live cell extrusion pathway and delamination of a subpopulation of NCC in parallel with EMT, which has implications for cell delamination in development and disease.
Collapse
Affiliation(s)
| | - Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS66160
| | | | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO64110
| | | | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS66160
| |
Collapse
|
34
|
Bautista GM, Du Y, Matthews MJ, Flores AM, Kushnir NR, Sweeney NK, Nguyen NPN, Tokhtaeva E, Solorzano-Vargas RS, Lewis M, Stelzner M, He X, Dunn JCY, Martin MG. Smooth muscle cell Piezo1 depletion results in impaired contractile properties in murine small bowel. Commun Biol 2025; 8:448. [PMID: 40097724 PMCID: PMC11914552 DOI: 10.1038/s42003-025-07697-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Piezo1 is a mechanosensitive cation channel expressed in intestinal muscularis cells (IMCs), including smooth muscle cells (SMCs), interstitial cells of Cajal, and Pdgfrα+ cells, which form the SIP syncytium, crucial for GI contractility. Here, we investigate the effects of SMC-specific Piezo1 deletion on small bowel function. Piezo1 depletion results in weight loss, delayed GI transit, muscularis thinning, and decreased SMCs. Ex vivo analyses demonstrated impaired contractile strength and tone, while in vitro studies using IMC co-cultures show dysrhythmic Ca2+ flux with decreased frequency. Imaging reveal that Piezo1 localizes intracellularly, thereby likely impacting Ca2+ signaling mechanisms modulated by Ca2 + -handling channels located on the sarcoplasmic reticulum and plasma membrane. Our findings suggest that Piezo1 in small bowel SMCs contributes to contractility by maintaining intracellular Ca2+ activity and subsequent signaling within the SIP syncytium. These findings provide new insights into the complex role of Piezo1 in small bowel SMCs and its implications for GI motility.
Collapse
Affiliation(s)
- Geoanna M Bautista
- Department of Pediatrics, Division of Neonatology, University of California Davis Children's Hospital, Sacramento, CA, 95817, USA
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yingjie Du
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Michael J Matthews
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Allison M Flores
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Nicole R Kushnir
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Nicolle K Sweeney
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Nam Phuong N Nguyen
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Elmira Tokhtaeva
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - R S Solorzano-Vargas
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Michael Lewis
- Department of Pathology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Matthias Stelzner
- Department of Surgery, VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Ximin He
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - James C Y Dunn
- Division of Pediatric Surgery, Departments of Surgery and Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Martin G Martin
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA.
| |
Collapse
|
35
|
Miao L, Lu Y, Nusrat A, Fan G, Zhang S, Zhao L, Wu CL, Guo H, Huyen TLN, Zheng Y, Fan ZC, Shou W, Schwartz RJ, Liu Y, Kumar A, Sui H, Serysheva II, Burns AR, Wan LQ, Zhou B, Evans SM, Wu M. Tunneling nanotube-like structures regulate distant cellular interactions during heart formation. Science 2025; 387:eadd3417. [PMID: 40080583 DOI: 10.1126/science.add3417] [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: 06/06/2022] [Revised: 08/26/2024] [Accepted: 01/03/2025] [Indexed: 03/15/2025]
Abstract
In the developing mammalian heart, the endocardium and the myocardium are separated by so-called cardiac jelly. Communication between the endocardium and the myocardium is essential for cardiac morphogenesis. How membrane-localized receptors and ligands achieve interaction across the cardiac jelly is not understood. Working in developing mouse cardiac morphogenesis models, we used a variety of cellular, imaging, and genetic approaches to elucidate this question. We found that myocardium and endocardium interacted directly through microstructures termed tunneling nanotube-like structures (TNTLs). TNTLs extended from cardiomyocytes (CMs) to contact endocardial cells (ECs) directly. TNTLs transported cytoplasmic proteins, transduced signals between CMs and ECs, and initiated myocardial growth toward the heart lumen to form ventricular trabeculae-like structures. Loss of TNTLs disturbed signaling interactions and, subsequently, ventricular patterning.
Collapse
Affiliation(s)
- Lianjie Miao
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Yangyang Lu
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Anika Nusrat
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Guizhen Fan
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shaohua Zhang
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Luqi Zhao
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Chia-Ling Wu
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Hongyan Guo
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Trang Le Nu Huyen
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, 3333 Brunet Avenue, Cincinnati, OH, USA
| | - Zhen-Chuan Fan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Weinian Shou
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Yu Liu
- Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Ashok Kumar
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Haixin Sui
- Division of Translational Medicine, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, USA
| | - Irina I Serysheva
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alan R Burns
- College of Optometry, University of Houston, Houston, TX, USA
| | - Leo Q Wan
- Departments of Biomedical Engineering and Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Bin Zhou
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Mingfu Wu
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| |
Collapse
|
36
|
Wang L, Tang Y, Buckley AF, Spurney RF. Podocyte specific knockout of the natriuretic peptide clearance receptor is podocyte protective in focal segmental glomerulosclerosis. PLoS One 2025; 20:e0319424. [PMID: 40063586 PMCID: PMC11892885 DOI: 10.1371/journal.pone.0319424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/31/2025] [Indexed: 05/13/2025] Open
Abstract
Natriuretic peptides (NPs) bind to glomerular podocytes and attenuate glomerular injury. The beneficial effects of NPs are negatively regulated by the NP clearance receptor (NPRC), which is highly expressed in podocytes. To determine if inhibiting NPRC is podocyte protective, we examined the effects of deleting NPRC in both cultured podocytes and in vivo. We found that: 1.Both atrial NP and C-type NP inhibit podocyte apoptosis in cultured podocytes, but these podocyte protective effects are significantly attenuated in cells expressing NPRC, and 2. Atrial NP was significantly more effective than CNP at inhibiting the apoptotic response. Consistent with the protective actions of NPs, podocyte specific knockout of NPRC reduced albuminuria, glomerular sclerosis and tubulointerstitial inflammation in a mouse model of focal segmental glomerulosclerosis. These beneficial actions were associated with: 1. Decreased expression of the myofibroblast marker alpha-smooth muscle actin, 2. Reduced expression of the extracellular matrix proteins collagen 4-alpha-1 and fibronectin, and 3. Preserved expression of the podocyte proteins nephrin and podocin. Inhibiting NP clearance may be a useful therapeutic approach to treat glomerular diseases.
Collapse
MESH Headings
- Podocytes/metabolism
- Podocytes/pathology
- Podocytes/drug effects
- Animals
- Glomerulosclerosis, Focal Segmental/pathology
- Glomerulosclerosis, Focal Segmental/metabolism
- Glomerulosclerosis, Focal Segmental/genetics
- Receptors, Atrial Natriuretic Factor/genetics
- Receptors, Atrial Natriuretic Factor/metabolism
- Receptors, Atrial Natriuretic Factor/deficiency
- Mice
- Apoptosis/drug effects
- Mice, Knockout
- Natriuretic Peptide, C-Type/pharmacology
- Natriuretic Peptide, C-Type/metabolism
- Disease Models, Animal
- Membrane Proteins/metabolism
- Albuminuria/genetics
- Male
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Liming Wang
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, United States of America
| | - Yuping Tang
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, United States of America
| | - Anne F. Buckley
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Robert F. Spurney
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, United States of America
| |
Collapse
|
37
|
McIntyre TI, Valdez O, Kochhar NP, Davidson B, Samad B, Qiu L, Hu K, Combes AJ, Erlebacher A. KDM6B-dependent epigenetic programming of uterine fibroblasts in early pregnancy regulates parturition timing in mice. Cell 2025; 188:1265-1279.e18. [PMID: 39842437 PMCID: PMC11890963 DOI: 10.1016/j.cell.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 09/03/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025]
Abstract
Current efforts investigating parturition timing mechanisms have focused on the proximal triggers of labor onset generated in late pregnancy. By studying the delayed parturition phenotype of mice with uterine fibroblast deficiencies in the histone H3K27me3 demethylase KDM6B, we provide evidence that parturition timing is regulated by events that take place in early pregnancy. Immediately after copulation, uterine fibroblasts engage in a locus-specific epigenetic program that abruptly adjusts H3K27me3 levels across their genome. In the absence of KDM6B, many of the adjusted loci over-accumulate H3K27me3. This over-accumulation leads to nearby genes being misexpressed in mid-to-late gestation, a delayed effect partly attributable to a second locus-specific but KDM6B-independent process initiated within uterine fibroblasts soon after implantation. This second process employs progressive H3K27me3 loss to temporally structure post-midgestational patterns of gene induction. Further dissection of the ways uterine programming controls parturition timing may have relevance to human pregnancy complications such as preterm labor.
Collapse
Affiliation(s)
- Tara I McIntyre
- Biomedical Sciences Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Omar Valdez
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nathan P Kochhar
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Brittany Davidson
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bushra Samad
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Longhui Qiu
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kenneth Hu
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexis J Combes
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Adrian Erlebacher
- Biomedical Sciences Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; Center for Reproductive Science, School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
38
|
Schoultz E, Moccia C, Liang S, Johansson E, Nilsson M. Tumor Cell Plasticity and Stromal Microenvironment Distinguish Papillary and Follicular Growth Patterns in a Mouse Model of BRAFV600E-Induced Thyroid Cancer. CANCER RESEARCH COMMUNICATIONS 2025; 5:409-421. [PMID: 39956582 PMCID: PMC11885905 DOI: 10.1158/2767-9764.crc-24-0474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/27/2024] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
SIGNIFICANCE Cell-of-origin intrinsic features rather than driver mutation identity influence tumor growth patterning in differentiated thyroid cancer and might impact histopathologic diagnosis of thyroid carcinoma subtypes.
Collapse
Affiliation(s)
- Elin Schoultz
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Carmen Moccia
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Shawn Liang
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Ellen Johansson
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Mikael Nilsson
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
- Department of Medical Chemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
39
|
Gao Z, Yu Y, Eckel‐Mahan K, Kolonin MG. Caloric Restriction and Telomere Preservation in TERT Knockout Adipocyte Progenitors Does Not Rescue Mice From Metabolic Dysfunction due to a TERT Function in Adipocyte Mitochondria. Aging Cell 2025; 24:e14499. [PMID: 39932851 PMCID: PMC11896407 DOI: 10.1111/acel.14499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/20/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
Inactivation of telomerase (TERT) in adipocyte progenitor cells (APC) expedites telomere attrition, and the onset of diabetes in mice fed high-fat diet (HFD), which promotes APC over-proliferation and replicative senescence. Here, we show that time-restricted feeding or caloric restriction in the postnatal development of mice subsequently subjected to HFD prevents telomere attrition but not glucose intolerance. This metabolic effect of dietary intervention was not observed for mice with TERT KO in endothelial or myeloid cells. To characterize the telomere-independent effects of TERT in the APC lineage, we analyzed mice with TERT knockout in mature adipocytes (AD-TERT-KO), which do not proliferate and avoid telomere attrition. Analysis of adipocytes from AD-TERT-KO mice indicated reliance on glycolysis and decreased mitochondrial oxidative metabolism. We show that AD-TERT-KO mice have reduced cold tolerance and metabolism abnormality indicating a defect in adaptive thermogenesis, characteristic of aging. Conversely, ectopic TERT expression in brown adipocytes-induced mitochondrial oxidation and thermogenic gene expression. We conclude that TERT plays an important non-canonical function in the mitochondria of adipocytes.
Collapse
Affiliation(s)
- Zhanguo Gao
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Kristin Eckel‐Mahan
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| |
Collapse
|
40
|
Moghe P, Belousov R, Ichikawa T, Iwatani C, Tsukiyama T, Erzberger A, Hiiragi T. Coupling of cell shape, matrix and tissue dynamics ensures embryonic patterning robustness. Nat Cell Biol 2025; 27:408-423. [PMID: 39966670 PMCID: PMC11906357 DOI: 10.1038/s41556-025-01618-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/20/2024] [Indexed: 02/20/2025]
Abstract
Tissue patterning coordinates morphogenesis, cell dynamics and fate specification. Understanding how precision in patterning is robustly achieved despite inherent developmental variability during mammalian embryogenesis remains a challenge. Here, based on cell dynamics quantification and simulation, we show how salt-and-pepper epiblast and primitive endoderm (PrE) cells pattern the inner cell mass of mouse blastocysts. Coupling cell fate and dynamics, PrE cells form apical polarity-dependent actin protrusions required for RAC1-dependent migration towards the surface of the fluid cavity, where PrE cells are trapped due to decreased tension. Concomitantly, PrE cells deposit an extracellular matrix gradient, presumably breaking the tissue-level symmetry and collectively guiding their own migration. Tissue size perturbations of mouse embryos and their comparison with monkey and human blastocysts further demonstrate that the fixed proportion of PrE/epiblast cells is optimal with respect to embryo size and tissue geometry and, despite variability, ensures patterning robustness during early mammalian development.
Collapse
Grants
- The Hiiragi laboratory was supported by the EMBL, and currently by the Hubrecht Institute, the European Research Council (ERC Advanced Grant “SelforganisingEmbryo” grant agreement 742732, ERC Advanced Grant “COORDINATION” grant agreement 101055287), Stichting LSH-TKI (LSHM21020), and Japan Society for the Promotion of Science (JSPS) KAKENHI grant numbers JP21H05038 and JP22H05166. The Erzberger laboratory is supported by the EMBL.
- European Molecular Biology Laboratory (EMBL Heidelberg)
- MEXT | Japan Society for the Promotion of Science (JSPS)
- T.I. was supported by the JSPS Overseas Research Fellowship
- The Erzberger laboratory is supported by the EMBL.
- The Hiiragi laboratory was supported by the EMBL, and currently by the Hubrecht Institute, the European Research Council (ERC Advanced Grant “SelforganisingEmbryo” grant agreement 742732, ERC Advanced Grant “COORDINATION” grant agreement 101055287), Stichting LSH-TKI (LSHM21020), and Japan Society for the Promotion of Science (JSPS) KAKENHI grant numbers JP21H05038 and JP22H05166.
Collapse
Affiliation(s)
- Prachiti Moghe
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Utrecht, Netherlands
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Roman Belousov
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Takafumi Ichikawa
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chizuru Iwatani
- Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, Japan
| | - Tomoyuki Tsukiyama
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, Japan
| | - Anna Erzberger
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Takashi Hiiragi
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Utrecht, Netherlands.
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan.
- Department of Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
41
|
Juros D, Hastings RL, Pendragon A, Kay J, Valdez G. Loss of MEGF10 Decreases the Number of Perisynaptic Schwann Cells and Innervation of Neuromuscular Junctions in Aging Mice. J Peripher Nerv Syst 2025; 30:e70014. [PMID: 40099661 DOI: 10.1111/jns.70014] [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: 06/21/2024] [Revised: 02/20/2025] [Accepted: 03/07/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND AND AIMS At the neuromuscular junction (NMJ), the synapse between motor neurons and muscle fibers, reside perisynaptic Schwann cells (PSCs) which are specialized glia that regulate the maintenance and repair of this synapse. While we know how PSC morphology and numbers change in aging and various neuromuscular disorders that adversely affect the NMJ, the molecular mechanisms that alter PSC functions remain unknown. In this study, we investigated whether MEGF10 in PSCs modulates NMJ stability in developing, healthy young adult, middle-aged, and axotomized mice. MEGF10 is a glial phagocytic receptor that is enriched in PSCs compared to other Schwann cells (SCs). METHODS We isolated PSCs from a transgenic reporter mouse line to assess Megf10 expression at different ages and following nerve injury using qPCR. We then used a conditional mouse lacking Megf10 in all SCs, including PSCs (Megf10 SC-KO mice). We examined NMJs and axonal debris clearance in Megf10 SC-KO mice using confocal microscopy. RESULTS We found that Megf10 expression in PSCs peaks during development and decreases during aging and following denervation of NMJs. NMJs were morphologically normal in developing and young adult Megf10 SC-KO mice. This was not the case in middle-aged Megf10 SC-KO mice, in which NMJs presented with fewer PSCs, decreased PSC coverage of the endplate, and decreased innervation in comparison to control mice. Following nerve injury-induced damage, axonal debris at the NMJ was cleared faster in Megf10 SC-KO mice; yet, the rate of reinnervation was unchanged compared to control mice. INTERPRETATION The data in this study suggest that MEGF10 in PSCs functions to maintain PSC number and NMJ innervation during aging. This study also suggests important roles for MEGF10 in mediating the clearance of axonal debris at NMJs following nerve injury.
Collapse
Affiliation(s)
- Devin Juros
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Robert Louis Hastings
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Ariane Pendragon
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jeremy Kay
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science, and Center on the Biology of Aging, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
42
|
Tauch S, Hey J, Kast B, Gengenbacher N, Weiß L, Sator‐Schmitt M, Lohr S, Brobeil A, Schirmacher P, Utikal J, Augustin HG, Plass C, Angel P. A Unique Signature for Cancer-Associated Fibroblasts in Melanoma Metastases. Pigment Cell Melanoma Res 2025; 38:e70002. [PMID: 39924882 PMCID: PMC11808227 DOI: 10.1111/pcmr.70002] [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: 07/03/2024] [Revised: 11/27/2024] [Accepted: 01/16/2025] [Indexed: 02/11/2025]
Abstract
Cancer-associated fibroblasts (CAFs) represent a central cell population of the tumor microenvironment (TME). Recently, single-cell RNA-sequencing (scRNA-seq) analyses of primary tumors of different cancer entities yielded different classifications of CAF subsets underscoring the heterogeneity of CAFs within the TME. Here, we analyzed the transcriptional signatures of approximately 8400 CAFs and normal fibroblasts by scRNA-seq and compared genetic profiles of CAFs from murine melanoma primary tumors to CAFs from corresponding melanoma lung metastases. This revealed distinct subsets for primary tumor and metastasis-specific CAF populations, respectively. Combined with the spatial characterization of metastasis CAFs at the RNA and protein level, scRNA analyses indicate tumor-dependent crosstalk between neutrophils and CAFs, mediated via SAA3 and IL1b-related signaling pathways, which can be recapitulated in vitro. Analyzing tissue sections of human patient samples, this interaction was found to be present in human melanoma metastasis. Taken together, our data highlight unique characteristics of metastasis CAFs with potential therapeutic impact for melanoma metastasis.
Collapse
Affiliation(s)
- Saskia Tauch
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Joschka Hey
- Division of Cancer EpigenomicsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Bettina Kast
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Nicolas Gengenbacher
- Division of Vascular Oncology and MetastasisGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
- European Center for Angioscience (ECAS), Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- DKFZ‐Hector Cancer Institute, University Medical Centre MannheimMannheimGermany
| | - Lena Weiß
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Melanie Sator‐Schmitt
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Sabrina Lohr
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Alexander Brobeil
- Institute of PathologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Peter Schirmacher
- Institute of PathologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Jochen Utikal
- DKFZ‐Hector Cancer Institute, University Medical Centre MannheimMannheimGermany
- Skin Cancer UnitGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Dermatology, Venereology and AllergologyUniversity Medical Center Mannheim, Ruprecht‐Karl University of HeidelbergMannheimGermany
| | - Hellmut G. Augustin
- Division of Vascular Oncology and MetastasisGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
- European Center for Angioscience (ECAS), Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- DKFZ‐Hector Cancer Institute, University Medical Centre MannheimMannheimGermany
| | - Christoph Plass
- Division of Cancer EpigenomicsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Peter Angel
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| |
Collapse
|
43
|
Chalifoux M, Avdeeva M, Posfai E. Geometric, cell cycle and maternal-to-zygotic transition-associated YAP dynamics during preimplantation embryo development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640568. [PMID: 40060487 PMCID: PMC11888467 DOI: 10.1101/2025.02.27.640568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
During the first cell fate decision in mammalian embryos the inner cell mass cells, which will give rise to the embryo proper and other extraembryonic tissues, segregate from the trophectoderm cells, the precursors of the placenta. Cell fate segregation proceeds in a gradual manner encompassing two rounds of cell division, as well as cell positional and morphological changes. While it is known that the activity of the Hippo signaling pathway and the subcellular localization of its downstream effector YAP dictate lineage specific gene expression, the response of YAP to these dynamic cellular changes remains incompletely understood. Here we address these questions by quantitative live imaging of endogenously tagged YAP while simultaneously monitoring geometric cellular features and cell cycle progression throughout cell fate segregation. We apply a probabilistic model to our dynamic data, providing a quantitative characterization of the mutual effects of YAP and cellular relative exposed area, which has previously been shown to correlate with subcellular YAP localization in fixed samples. Additionally, we study how nuclear YAP levels are influenced by other factors, such as the decreasing pool of maternally provided YAP that is partitioned to daughter cells through cleavage divisions, cell cycle-associated nuclear volume changes, and a delay after divisions in adjusting YAP levels to new cell positions. Interestingly, we find that establishing low nuclear YAP levels required for the inner cell mass fate is largely achieved by passive cell cycle-associated mechanisms. Moreover, contrary to expectations, we find that mechanical perturbations that result in cell shape changes do not influence YAP localization in the embryo. Together our work identifies how various inputs are integrated over a dynamic developmental time course to shape the levels of a key molecular determinant of the first cell fate choice.
Collapse
Affiliation(s)
- Madeleine Chalifoux
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, USA
| | - Maria Avdeeva
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
44
|
Dang K, Singh A, Chen X, Cotton JL, Guo S, Hu X, Tao Z, Liu H, Zhu LJ, Ip YT, Wu X, Mao J. Mesenchymal Hippo signaling regulates intestinal homeostasis in adult mice. iScience 2025; 28:111847. [PMID: 39981512 PMCID: PMC11841074 DOI: 10.1016/j.isci.2025.111847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/16/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
Intestinal homeostasis is tightly regulated by the reciprocal interaction between the gut epithelium and adjacent mesenchyme. The Hippo pathway is intimately associated with intestinal epithelial homeostasis and regeneration; however, its role in postnatal gut mesenchyme remains poorly defined. Here, we find that removal of the core Hippo kinases Lats1/2 or activation of YAP in adult intestinal smooth muscle layers has largely no effect; however, Hippo-YAP signaling in the niche-forming Gli1+ mesenchymal cells plays intrinsic roles in regulating intestinal homeostasis. We find that Lats1/2 deletion drives robust mesenchymal over-proliferation, and YAP activation in Gli1+ pericryptal cells disrupts the intestinal epithelial-mesenchymal crosstalk via promoting Wnt ligand production. We show that YAP is upregulated in the stroma during dextran sodium sulfate (DSS)-induced injury, and mesenchymal YAP activation facilitates intestinal epithelial regeneration. Altogether, our data suggest an important role for mesenchymal Hippo-YAP signaling in the stem cell niche during intestinal homeostasis and pathogenesis.
Collapse
Affiliation(s)
- Kyvan Dang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Alka Singh
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Xin Chen
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jennifer L. Cotton
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Susu Guo
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Xiaodi Hu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Nutrition and Food Sciences, Texas Woman’s University, Denton, TX 76204, USA
| | - Haibo Liu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Lihua J. Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Y. Tony Ip
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Xu Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
45
|
Biggs O'May J, Vanes L, de Boer LL, Lewis DA, Hartweger H, Kunzelmann S, Hayward D, Llorian M, Köchl R, Tybulewicz VLJ. WNK1-dependent water influx is required for CD4 + T cell activation and T cell-dependent antibody responses. Nat Commun 2025; 16:1857. [PMID: 39984435 PMCID: PMC11845700 DOI: 10.1038/s41467-025-56778-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/29/2025] [Indexed: 02/23/2025] Open
Abstract
Signaling from the T cell antigen receptor (TCR) on CD4+ T cells plays a critical role in adaptive immune responses by inducing T cell activation, proliferation, and differentiation. Here we demonstrate that WNK1, a kinase implicated in osmoregulation in the kidney, is required in T cells to support T-dependent antibody responses. We show that the canonical WNK1-OXSR1-STK39 kinase signaling pathway is required for TCR signaling in CD4+ T cells, their subsequent entry into the cell cycle, and suppression of the ATR-mediated G2/M cell cycle checkpoint. We show that the WNK1 pathway regulates ion influx leading to water influx, potentially through AQP3, and that water influx is required for TCR-induced signaling and cell cycle entry. Thus, TCR signaling via WNK1, OXSR1, STK39 and AQP3 leads to water entry that is essential for CD4+ T cell proliferation and hence T cell-dependent antibody responses.
Collapse
Affiliation(s)
| | - Lesley Vanes
- The Francis Crick Institute, London, NW1 1AT, UK
| | - Leonard L de Boer
- The Francis Crick Institute, London, NW1 1AT, UK
- Imperial College, London, W12 0NN, UK
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institute, Box 1031, SE-171 21, Solna, Sweden
| | | | - Harald Hartweger
- The Francis Crick Institute, London, NW1 1AT, UK
- Laboratory of Molecular Immunology, The Rockefeller University, 10065, New York, NY, USA
| | | | - Darryl Hayward
- The Francis Crick Institute, London, NW1 1AT, UK
- GSK, Stevenage, SG1 2NY, UK
| | | | - Robert Köchl
- The Francis Crick Institute, London, NW1 1AT, UK
- Kings College London, London, SE1 9RT, UK
| | | |
Collapse
|
46
|
Bermudez A, Latham ZD, Ma AJ, Bi D, Hu JK, Lin NYC. Regulation of chromatin modifications through coordination of nucleus size and epithelial cell morphology heterogeneity. Commun Biol 2025; 8:269. [PMID: 39979587 PMCID: PMC11842846 DOI: 10.1038/s42003-025-07677-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: 04/18/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
Cell morphology heterogeneity is pervasive in epithelial collectives, yet the underlying mechanisms driving such heterogeneity and its consequential biological ramifications remain elusive. Here, we observed a consistent correlation between the epithelial cell morphology and nucleus morphology during crowding, revealing a persistent log-normal probability distribution characterizing both cell and nucleus areas across diverse epithelial model systems. We showed that this morphological diversity arises from asymmetric partitioning during cell division. Next, we provide insights into the impact of nucleus morphology on chromatin modifications. We demonstrated that constraining nucleus leads to downregulation of the euchromatic mark H3K9ac and upregulation of the heterochromatic mark H3K27me3. Furthermore, we showed that nucleus size regulates H3K27me3 levels through histone demethylase UTX. These findings highlight the significance of cell morphology heterogeneity as a driver of chromatin state diversity, shaping functional variability within epithelial tissues.
Collapse
Affiliation(s)
- Alexandra Bermudez
- Bioengineering Department, University of California Los Angeles, Los Angeles, CA, USA
| | - Zoe D Latham
- Bioengineering Department, University of California Los Angeles, Los Angeles, CA, USA
| | - Alex J Ma
- Bioengineering Department, University of California Los Angeles, Los Angeles, CA, USA
| | - Dapeng Bi
- Department of Physics, Northeastern University, Boston, MA, USA
| | - Jimmy K Hu
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Broad Stem Cell Center, University of California Los Angeles, Los Angeles, CA, USA.
| | - Neil Y C Lin
- Bioengineering Department, University of California Los Angeles, Los Angeles, CA, USA.
- Broad Stem Cell Center, University of California Los Angeles, Los Angeles, CA, USA.
- Mechanical and Aerospace Engineering Department, University of California Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA.
- Institute for Quantitative and Computational Biosciences, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
47
|
Álvarez-Aznar A, Desai M, Orlich MM, Vázquez-Liébanas E, Adams RH, Brakebusch C, Gaengel K. Cdc42 is crucial for mural cell migration, proliferation and patterning of the retinal vasculature. Vascul Pharmacol 2025; 159:107472. [PMID: 39971261 DOI: 10.1016/j.vph.2025.107472] [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: 09/30/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
AIMS Mural cells constitute the outer lining of blood vessels and are essential for vascular development and function. Mural cell loss or malfunction has been associated with numerous diseases including diabetic retinopathy, stroke and amyotrophic lateral sclerosis. In this work, we investigate the role of CDC42 in mural cells in vivo, using the developing mouse retina as a model. METHODS In this study, we generated a mouse model for Cdc42 deletion in mural cells by crossing Pdgfrb-CreERT2 mice with Cdc42flox/flox mice. This model (Cdc42iΔMC) allowed us to investigate the role of CDC42 in pericytes and smooth muscle cells in the developing and adult retinal vasculature. RESULTS We find that, during postnatal development, CDC42 is required in both, pericytes and smooth muscle cells to maintain proper cell morphology, mural cell coverage and distribution. During retinal angiogenesis, Cdc42-depleted pericytes lag behind the sprouting front and exhibit decreased proliferation. Consequently, capillaries at the sprouting front remain pericyte deprived, become dilated and are prone to increased vascular leakage. In addition, arteries and arterioles deviate from their normal growth directions and trajectory. While in the adult retina, mural cell coverage normalizes and pericytes adopt a normal morphology, smooth muscle cell morphologies remain abnormal and arteriolar branching angles are markedly reduced. CONCLUSIONS Our findings demonstrate that CDC42 is required for mural cell migration and proliferation and suggest that mural cells are essential for normal morphogenesis and patterning of the developing retinal vasculature.
Collapse
Affiliation(s)
- Alberto Álvarez-Aznar
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Malavika Desai
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Michael M Orlich
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elisa Vázquez-Liébanas
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Ralf H Adams
- Max Plank Institute for Molecular Medicine, Department of Tissue Morphogenesis, Münster, Germany
| | | | - Konstantin Gaengel
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
48
|
Bermudez A, Latham ZD, Ma AJ, Bi D, Hu JK, Lin NYC. Regulation of Chromatin Modifications through Coordination of Nucleus Size and Epithelial Cell Morphology Heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.18.590164. [PMID: 38712099 PMCID: PMC11071433 DOI: 10.1101/2024.04.18.590164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Cell morphology heterogeneity is pervasive in epithelial collectives, yet the underlying mechanisms driving such heterogeneity and its consequential biological ramifications remain elusive. Here, we observed a consistent correlation between the epithelial cell morphology and nucleus morphology during crowding, revealing a persistent log-normal probability distribution characterizing both cell and nucleus areas across diverse epithelial model systems. We further showed that this morphological diversity arises from asymmetric partitioning during cell division. Moreover, we provide insights into the impact of nucleus morphology on chromatin modifications. We demonstrated that constraining nucleus leads to downregulation of the euchromatic mark H3K9ac and upregulation of the heterochromatic mark H3K27me3. Furthermore, we showed that nucleus size regulates H3K27me3 levels through histone demethylase UTX. These findings highlight the significance of cell morphology heterogeneity as a driver of chromatin state diversity, shaping functional variability within epithelial tissues.
Collapse
|
49
|
Luna G, Verheyden J, Tan C, Kim E, Hwa M, Sahi J, Shen Y, Chung W, McCulley D, Sun X. MYRF is Essential in Mesothelial Cells to Promote Lung Development and Maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.635155. [PMID: 39990361 PMCID: PMC11844445 DOI: 10.1101/2025.02.13.635155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The mesothelium is a squamous monolayer that ensheathes internal organs, lines the body cavity, and the diaphragm. It serves as a protective barrier, coated in glycocalyx, and secretes lubricants to facilitate tissue movement. How the mesothelium forms is poorly understood. Here, we investigate Myrf , a transcription factor gene expressed in the mesothelium, because it carries variants in patients with Congenital Diaphragmatic Hernia (CDH), a disorder that affects the diaphragm, lung, and other organs. In mice, inactivation of Myrf early in organogenesis resulted in CDH and defective mesothelial specification, compromising its function as a signaling center for lung growth. Inactivation of Myrf later led to enhanced mesothelium differentiation into mesenchymal cell types through partial epithelial-to-mesenchymal transition (EMT), resulting in a unique accumulation of smooth muscle encasing the lung. In this role, MYRF functions in parallel with YAP/TAZ. Together, these findings establish MYRF as a critical regulator of mesothelium development, and when mutated, causes CDH.
Collapse
|
50
|
Chugh RM, Bhanja P, Zitter R, Gunewardena S, Badkul R, Saha S. Modulation of β-Catenin promotes WNT expression in macrophages and mitigates intestinal injury. Cell Commun Signal 2025; 23:78. [PMID: 39934819 PMCID: PMC11818365 DOI: 10.1186/s12964-025-02065-7] [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: 10/10/2024] [Accepted: 01/25/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Macrophages are the major source of WNT ligands. However, the regulation of WNT expression in macrophages has not been studied. In the present study, we have discovered that activation of canonical β-Catenin signaling suppresses WNT expression in macrophages. EVs from these pre-conditioned macrophages promoted intestinal stem cell regeneration and mitigated intestinal injury. METHOD ChIP-seq analysis and validation studies using recombinant DNA construct expressing Luciferase reporter under WNT promoter (e.g. WNT5a and WNT9b) were conducted to demonstrate the involvement of β-Catenin in the transcriptional regulation of WNT expression. The regulatory role of β-Catenin in WNT expression in macrophages was examined by treating these cells with a Tankyrase inhibitor. In addition, the gene expressing β-Catenin was deleted in macrophages using Csf1r.iCre; Ctnnb1fl/fl mice model. Both pharmacological and genetically modulated macrophages were examined for WNT expression and activity by qPCR and TCF/LEF luciferase assay respectively. Additionally, Csf1r.iCre; Ctnnb1fl/fl mice were exposed to irradiation to compare the radiosensitivity with their wildtype littermate. Extracellular vesicles (EVs) were isolated from pre-conditioned WNT-enriched macrophages and infused in irradiated C57BL/6 and Lgr5/eGFP-IRES-Cre-ERT2; R26-ACTB-tdTomato-EGFP mice to determine the regenerative response of intestinal stem cell (ISC) and epithelial repair. Regenerative effects of EVs were also examined in mice model DSS induced colitis. RESULT ChIP-seq analysis and subsequent validation study suggested physical association of β-Catenin with WNT promoters to suppress WNT expression. Macrophage specific deletion of gene expressing β-Catenin or pharmacological inhibition of Tankyrase improves the WNT expression in macrophages several folds compared to control. Transfusion of these preconditioned macrophages or EVs from these cells delivers optimum level of morphogenic WNT to injured epithelium, activates ISC regeneration and mitigated radiation induced intestinal injury. Intestinal epithelium in Csf1r.iCre; Ctnnb1fl/fl mice also showed radioresistance compared to wild type littermate. Moreover, EVs derived from WNT enriched macrophages can mitigate intestinal injury in mice model of DSS induced acute colitis. CONCLUSION The study provides substantial evidence that macrophage-targeted modulation of canonical WNT signaling induces WNT expression in macrophages. Treatment with preconditioned macrophage derived WNT-enriched EVs can be a promising therapeutic approach against intestinal injury.
Collapse
Affiliation(s)
- Rishi Man Chugh
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Payel Bhanja
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ryan Zitter
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Sumedha Gunewardena
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Rajeev Badkul
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Subhrajit Saha
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|