1
|
Nawaito SA, Esmael M, Souchkova O, Cardinal T, Bernas G, Bergeron K, Gayda F, Bergeron F, Bouchard M, Zhou X, Ongaro L, Bernard DJ, Short J, Wray S, Viger RS, Mounier C, Pilon N. Brain-Specific Gata4 Downregulation in Greywick Female Mice Models the Metabolic Subtype of Polycystic Ovary Syndrome. FASEB J 2025; 39:e70717. [PMID: 40515561 PMCID: PMC12166494 DOI: 10.1096/fj.202401718rr] [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: 07/25/2024] [Revised: 05/28/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogenous disorder characterized by reproductive and metabolic abnormalities. PCOS etiology remains poorly understood, although the hypothalamus is suspected to play a central role in many cases. Human genetic studies have also shown an association with the transcription factor-coding gene GATA4, but without providing a functional link. Here, we show that adult Greywick female mice may bridge this gap. These mice phenocopy PCOS with partial penetrance, due to the serendipitous insertion of a Gata4 promoter-driven transgene in a strong enhancer region. Resulting robust transgene expression in subsets of hypothalamic neurons and glia impairs endogenous Gata4 expression, resulting in misexpression of genes linked to the control of fertility and food intake. We also show that this previously overlooked role of GATA4 in the hypothalamus can be replicated by conditional knockout approaches. Overall, this study sheds light not only on PCOS etiology but also on the role played by GATA4 in the central control of reproduction.
Collapse
Affiliation(s)
- Sherin A. Nawaito
- Molecular Genetics of Development Laboratory, Department of Biological SciencesUniversité du Québec à MontréalMontrealCanada
- Centre d'Excellence en Recherche sur les Maladies Orphelines – Fondation CourtoisUniversité du Québec à MontréalMontrealCanada
- Department of PhysiologySuez Canal UniversityIsmailiaEgypt
| | - Mostafa Esmael
- Molecular Genetics of Development Laboratory, Department of Biological SciencesUniversité du Québec à MontréalMontrealCanada
- Centre d'Excellence en Recherche sur les Maladies Orphelines – Fondation CourtoisUniversité du Québec à MontréalMontrealCanada
| | - Ouliana Souchkova
- Molecular Genetics of Development Laboratory, Department of Biological SciencesUniversité du Québec à MontréalMontrealCanada
- Centre d'Excellence en Recherche sur les Maladies Orphelines – Fondation CourtoisUniversité du Québec à MontréalMontrealCanada
| | - Tatiana Cardinal
- Molecular Genetics of Development Laboratory, Department of Biological SciencesUniversité du Québec à MontréalMontrealCanada
- Centre d'Excellence en Recherche sur les Maladies Orphelines – Fondation CourtoisUniversité du Québec à MontréalMontrealCanada
| | - Guillaume Bernas
- Molecular Genetics of Development Laboratory, Department of Biological SciencesUniversité du Québec à MontréalMontrealCanada
- Centre d'Excellence en Recherche sur les Maladies Orphelines – Fondation CourtoisUniversité du Québec à MontréalMontrealCanada
| | - Karl‐F. Bergeron
- Centre d'Excellence en Recherche sur les Maladies Orphelines – Fondation CourtoisUniversité du Québec à MontréalMontrealCanada
- Lipid Metabolism Laboratory, Department of Biological SciencesUniversité du Québec à MontréalMontrealCanada
| | - Fanny Gayda
- Molecular Genetics of Development Laboratory, Department of Biological SciencesUniversité du Québec à MontréalMontrealCanada
| | - Francis Bergeron
- Reproduction, Mother and Child Health, Centre de recherche du CHU de QuébecUniversité LavalQuébecCanada
| | - Marie‐France Bouchard
- Reproduction, Mother and Child Health, Centre de recherche du CHU de QuébecUniversité LavalQuébecCanada
| | - Xiang Zhou
- Department of Pharmacology and TherapeuticsMcGill UniversityMontrealCanada
| | - Luisina Ongaro
- Department of Pharmacology and TherapeuticsMcGill UniversityMontrealCanada
| | - Daniel J. Bernard
- Department of Pharmacology and TherapeuticsMcGill UniversityMontrealCanada
| | - Jacob Short
- Cellular and Developmental Neurobiology SectionNational Institute of Neurological Disorders and StrokeBethesdaMarylandUSA
| | - Susan Wray
- Cellular and Developmental Neurobiology SectionNational Institute of Neurological Disorders and StrokeBethesdaMarylandUSA
| | - Robert S. Viger
- Reproduction, Mother and Child Health, Centre de recherche du CHU de QuébecUniversité LavalQuébecCanada
- Department of Obstetrics, Gynecology, and Reproduction, Centre de Recherche en Reproduction, Développement et Santé IntergénérationnelleUniversité LavalQuébecCanada
| | - Catherine Mounier
- Centre d'Excellence en Recherche sur les Maladies Orphelines – Fondation CourtoisUniversité du Québec à MontréalMontrealCanada
- Lipid Metabolism Laboratory, Department of Biological SciencesUniversité du Québec à MontréalMontrealCanada
| | - Nicolas Pilon
- Molecular Genetics of Development Laboratory, Department of Biological SciencesUniversité du Québec à MontréalMontrealCanada
- Centre d'Excellence en Recherche sur les Maladies Orphelines – Fondation CourtoisUniversité du Québec à MontréalMontrealCanada
- Department of PediatricsUniversité de MontréalMontrealCanada
| |
Collapse
|
2
|
Novak JSS, Polak L, Baksh SC, Barrows DW, Schernthanner M, Jackson BT, Thompson EAN, Gola A, Abdusselamoglu MD, Bonny AR, Gonzales KAU, Brunner JS, Bridgeman AE, Stewart KS, Hidalgo L, Dela Cruz-Racelis J, Luo JD, Gur-Cohen S, Pasolli HA, Carroll TS, Finley LWS, Fuchs E. The integrated stress response fine-tunes stem cell fate decisions upon serine deprivation and tissue injury. Cell Metab 2025:S1550-4131(25)00266-9. [PMID: 40513561 DOI: 10.1016/j.cmet.2025.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 03/11/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025]
Abstract
Epidermal stem cells produce the skin's barrier that excludes pathogens and prevents dehydration. Hair follicle stem cells (HFSCs) are dedicated to bursts of hair regeneration, but upon injury, they can also reconstruct, and thereafter maintain, the overlying epidermis. How HFSCs balance these fate choices to restore physiologic function to damaged tissue remains poorly understood. Here, we uncover serine as an unconventional, non-essential amino acid that impacts this process. When dietary serine dips, endogenous biosynthesis in HFSCs fails to meet demands (and vice versa), slowing hair cycle entry. Serine deprivation also alters wound repair, further delaying hair regeneration while accelerating re-epithelialization kinetics. Mechanistically, we show that HFSCs sense each fitness challenge by triggering the integrated stress response, which acts as a rheostat of epidermal-HF identity. As stress levels rise, skin barrier restoration kinetics accelerate while hair growth is delayed. Our findings offer potential for dietary and pharmacological intervention to accelerate wound healing.
Collapse
Affiliation(s)
- Jesse S S Novak
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Lisa Polak
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Sanjeethan C Baksh
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Douglas W Barrows
- Bioinformatics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Marina Schernthanner
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Benjamin T Jackson
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elizabeth A N Thompson
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Anita Gola
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - M Deniz Abdusselamoglu
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Alain R Bonny
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Kevin A U Gonzales
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Julia S Brunner
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna E Bridgeman
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Katie S Stewart
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Lynette Hidalgo
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - June Dela Cruz-Racelis
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Ji-Dung Luo
- Bioinformatics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Shiri Gur-Cohen
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - H Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Thomas S Carroll
- Bioinformatics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Lydia W S Finley
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
3
|
Gomez AR, Byun HR, Wu S, Muhammad AKMG, Ikbariyeh J, Chen J, Muro A, Li L, Bernstein KE, Ainsworth R, Tourtellotte WG. Boosting angiotensin-converting enzyme (ACE) in microglia protects against Alzheimer's disease in 5xFAD mice. NATURE AGING 2025:10.1038/s43587-025-00879-1. [PMID: 40490625 DOI: 10.1038/s43587-025-00879-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 04/17/2025] [Indexed: 06/11/2025]
Abstract
Genome-wide association studies have identified many gene polymorphisms associated with an increased risk of developing late-onset Alzheimer's disease (LOAD). Many of these LOAD risk-associated alleles alter disease pathogenesis by influencing innate immune responses and lipid metabolism of microglia (MG). Here we show that boosting the expression of angiotensin-converting enzyme (ACE), a genome-wide association study LOAD risk-associated gene product, specifically in MG, reduces amyloid-β (Aβ) plaque load, preserves vulnerable neurons and excitatory synapses, and significantly reduces learning and memory abnormalities in the 5xFAD amyloid mouse model of AD. ACE-expressing MG surround plaques more frequently and they have increased Aβ phagocytosis, endolysosomal trafficking and spleen tyrosine kinase activation downstream of the major Aβ receptors, triggering receptor expressed on myeloid cells 2 (Trem2) and C-type lectin domain family 7 member A (Clec7a). These findings establish a role for ACE in enhancing microglial immune function and they identify a potential use for ACE-expressing MG as a cell-based therapy to augment endogenous microglial responses to Aβ in AD.
Collapse
Affiliation(s)
- Andrew R Gomez
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hyae Ran Byun
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shaogen Wu
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - A K M Ghulam Muhammad
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jasmine Ikbariyeh
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jaelin Chen
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alek Muro
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lin Li
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kenneth E Bernstein
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Richard Ainsworth
- Kao Autoimmunity Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Warren G Tourtellotte
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Bhattacharya S, Tie G, Singh PNP, Malagola E, Eskiocak O, He R, Kraiczy J, Gu W, Perlov Y, Alici-Garipcan A, Beyaz S, Wang TC, Zhou Q, Shivdasani RA. Intestinal secretory differentiation reflects niche-driven phenotypic and epigenetic plasticity of a common signal-responsive terminal cell. Cell Stem Cell 2025; 32:952-969.e8. [PMID: 40203837 PMCID: PMC12145258 DOI: 10.1016/j.stem.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/27/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025]
Abstract
Enterocytes and four classic secretory cell types derive from intestinal epithelial stem cells. Based on morphology, location, and canonical markers, goblet and Paneth cells are considered distinct secretory types. Here, we report high overlap in their transcripts and sites of accessible chromatin, in marked contrast to those of their enteroendocrine or tuft cell siblings. Mouse and human goblet and Paneth cells express extraordinary fractions of few antimicrobial genes, which reflect specific responses to local niches. Wnt signaling retains some ATOH1+ secretory cells in crypt bottoms, where the absence of BMP signaling potently induces Paneth features. Cells that migrate away from crypt bottoms encounter BMPs and thereby acquire goblet properties. These phenotypes and underlying accessible cis-elements interconvert in post-mitotic cells. Thus, goblet and Paneth properties represent alternative phenotypic manifestations of a common signal-responsive terminal cell type. These findings reveal exquisite niche-dependent cell plasticity and cis-regulatory dynamics in likely response to antimicrobial needs.
Collapse
Affiliation(s)
- Swarnabh Bhattacharya
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Guodong Tie
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Pratik N P Singh
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Onur Eskiocak
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Graduate Program in Genetics, State University of New York, Stony Brook, NY 11794, USA
| | - Ruiyang He
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Judith Kraiczy
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Gu
- Division of Regenerative Medicine & Hartman Institute for Therapeutic Organ Regeneration, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yakov Perlov
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Qiao Zhou
- Division of Regenerative Medicine & Hartman Institute for Therapeutic Organ Regeneration, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
5
|
Torii S, Nagaharu K, Nakanishi N, Usui H, Hori Y, Hirose K, Toyosawa S, Morii E, Narushima M, Kubota Y, Nakagawa O, Imanaka-Yoshida K, Maruyama K. Embryological cellular origins and hypoxia-mediated mechanisms in PIK3CA-driven refractory vascular malformations. EMBO Mol Med 2025; 17:1289-1324. [PMID: 40234712 PMCID: PMC12162881 DOI: 10.1038/s44321-025-00235-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/17/2025] Open
Abstract
Congenital vascular malformations, affecting 0.5% of the population, often occur in the head and neck, complicating treatment due to the critical functions in these regions. Our previous research identified distinct developmental origins for blood and lymphatic vessels in these areas, tracing them to the cardiopharyngeal mesoderm (CPM), which contributes to the development of the head, neck, and cardiovascular system in both mouse and human embryos. In this study, we investigated the pathogenesis of these malformations by expressing Pik3caH1047R in the CPM. Mice expressing Pik3caH1047R in the CPM developed vascular abnormalities restricted to the head and neck. Single-cell RNA sequencing revealed that Pik3caH1047R upregulates Vegf-a expression in endothelial cells through HIF-mediated hypoxia signaling. Human samples supported these findings, showing elevated HIF-1α and VEGF-A in malformed vessels. Notably, inhibition of HIF-1α and VEGF-A in the mouse model significantly reduced abnormal vasculature. These results highlight the role of embryonic origins and hypoxia-driven mechanisms in vascular malformations, providing a foundation for the development of therapies targeting these difficult-to-treat conditions.
Collapse
Affiliation(s)
- Sota Torii
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Keiki Nagaharu
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan
| | - Nanako Nakanishi
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Hidehito Usui
- Department of Surgery, Kanagawa Children's Medical Center, 2-138-4, Mutsukawa, Minami-ku, Yokohama, Kanagawa, Japan
| | - Yumiko Hori
- Department of Pathology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Central Laboratory and Surgical Pathology, NHO Osaka National Hospital, 2-1-14 Hoenzaka, Chuo-ku, Osaka, 540-0006, Japan
| | - Katsutoshi Hirose
- Department of Oral and Maxillofacial Pathology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoru Toyosawa
- Department of Oral and Maxillofacial Pathology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mitsunaga Narushima
- Department of Plastic and Reconstructive Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Osamu Nakagawa
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-shimmachi, Suita, Osaka, 564-8565, Japan
| | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Kazuaki Maruyama
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
6
|
Sheridan D, Chakravarty P, Golan G, Shiakola Y, Olsen J, Burnett E, Galichet C, Fiordelisio T, Mollard P, Melamed P, Lovell-Badge R, Rizzoti K. Gonadotrophs have a dual origin, with most derived from early postnatal pituitary stem cells. Nat Commun 2025; 16:4280. [PMID: 40399281 PMCID: PMC12095566 DOI: 10.1038/s41467-025-59495-7] [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: 10/07/2024] [Accepted: 04/24/2025] [Indexed: 05/23/2025] Open
Abstract
Gonadotrophs are the essential pituitary endocrine cells for reproduction. They produce both luteinizing (LH) and follicle-stimulating (FSH) hormones that act on the gonads to promote germ cell maturation and steroidogenesis. Their secretion is controlled by the hypothalamic gonadotrophin-releasing hormone (GnRH), and gonadal steroid feedback. Gonadotrophs first appear in the embryonic pituitary, along with other endocrine cell types, and all expand after birth. While gonadotrophs may display heterogeneity in their response to GnRH, they appear, at least transcriptionally, as a homogenous population. The pituitary also contains a population of stem cells (SCs), whose contribution to postnatal growth is unclear, in part because endocrine cells maintain the ability to proliferate. Here we show an unsuspected dual origin of the murine adult gonadotroph population, with most gonadotrophs originating from postnatal pituitary stem cells starting early postnatally and up to puberty, while embryonic gonadotrophs are maintained. We further demonstrate that postnatal gonadotroph differentiation happens independently of gonadal signals and is not affected by impairment of GnRH signalling. The division of gonadotrophs based on separate origins has implications for our understanding of the establishment and regulation of reproductive functions, both in health and in disease.
Collapse
Affiliation(s)
- Daniel Sheridan
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, NW1 1AT, UK
| | | | - Gil Golan
- Faculty of Biology, Technion Israel Institute of Technology, Haifa, 32000, Israel
| | - Yiolanda Shiakola
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, NW1 1AT, UK
| | - Jessica Olsen
- Genetic Modification Service, The Francis Crick Institute, London, NW1 1AT, UK
| | - Elise Burnett
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, NW1 1AT, UK
| | - Christophe Galichet
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, NW1 1AT, UK
| | - Tatiana Fiordelisio
- Laboratorio de Neuroendocrinologia Comparada, Laboratorio Nacional de Soluciones Biomimeticas para Diagnostico y Terapia, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Patrice Mollard
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, Inserm, 34094, Montpellier, France
| | - Philippa Melamed
- Faculty of Biology, Technion Israel Institute of Technology, Haifa, 32000, Israel
| | - Robin Lovell-Badge
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, NW1 1AT, UK.
| | - Karine Rizzoti
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, NW1 1AT, UK.
| |
Collapse
|
7
|
Bastos J, O'Brien C, Vara-Pérez M, Mampay M, van Olst L, Barry-Carroll L, Kancheva D, Leduc S, Lievens AL, Ali L, Vlasov V, Meysman L, Shakeri H, Roelandt R, Van Hove H, De Vlaminck K, Scheyltjens I, Yaqoob F, Lombroso SI, Breugelmans M, Faron G, Gomez-Nicola D, Gate D, Bennett FC, Movahedi K. Monocytes can efficiently replace all brain macrophages and fetal liver monocytes can generate bona fide SALL1 + microglia. Immunity 2025; 58:1269-1288.e12. [PMID: 40311613 PMCID: PMC12094688 DOI: 10.1016/j.immuni.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
Microglia and border-associated macrophages (BAMs) are critical for brain health, and their dysfunction is associated to disease. Replacing brain macrophages holds substantial therapeutic promise but remains challenging. Here, we demonstrate that monocytes can efficiently replace all brain macrophages. Monocytes readily replaced embryonal BAMs upon their depletion and engrafted as monocyte-derived microglia (Mo-Microglia) upon more sustained niche availability. Mo-Microglia expanded comparably to their embryonic counterparts and showed similar longevity. However, monocytes were unable to replicate the distinct identity of embryonically derived BAMs and microglia. Using xenotransplantation, we found that human monocytes exhibited similar behavior, enabling identification of putative Mo-Microglia in Alzheimer's disease individuals. In mice and humans, monocyte ontogeny shaped their identity as brain macrophages. Importantly, mouse fetal liver monocytes exhibited a distinct epigenetic landscape and could develop a bona fide microglial identity. Our results illuminate brain macrophage development and highlight monocytes as an abundant progenitor source for brain macrophage replacement therapies.
Collapse
Affiliation(s)
- Jonathan Bastos
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carleigh O'Brien
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mónica Vara-Pérez
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Myrthe Mampay
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lynn van Olst
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Liam Barry-Carroll
- School of Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK; Nutrineuro, UMR 1286 INRAE, Bordeaux University, Bordeaux INP, Bordeaux, France
| | - Daliya Kancheva
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sophia Leduc
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ayla Line Lievens
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Leen Ali
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Vladislav Vlasov
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laura Meysman
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hadis Shakeri
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ria Roelandt
- VIB Single Cell Core, VIB, Ghent/Leuven, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Hannah Van Hove
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karen De Vlaminck
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Isabelle Scheyltjens
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fazeela Yaqoob
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sonia I Lombroso
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria Breugelmans
- Department of Obstetrics and Prenatal Medicine, UZ Brussel, VUB, Brussels, Belgium
| | - Gilles Faron
- Department of Obstetrics and Prenatal Medicine, UZ Brussel, VUB, Brussels, Belgium
| | - Diego Gomez-Nicola
- School of Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK
| | - David Gate
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - F Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kiavash Movahedi
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
8
|
Sussman JH, Cure HW, Yuan S, Ito K, Asangani IA, Garcia BA, Stanger BZ, Katsuda T. In vivo CRISPR screening reveals epigenetic regulators of hepatobiliary plasticity. Genes Dev 2025; 39:603-616. [PMID: 40169232 PMCID: PMC12047657 DOI: 10.1101/gad.352420.124] [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: 10/29/2024] [Accepted: 02/26/2025] [Indexed: 04/03/2025]
Abstract
Following prolonged liver injury, a small fraction of hepatocytes undergoes reprogramming to become cholangiocytes or biliary epithelial cells (BECs). This physiological process involves chromatin and transcriptional remodeling, but the epigenetic mediators are largely unknown. Here, we exploited a lineage-traced model of liver injury to investigate the role of histone post-translational modification in biliary reprogramming. Using mass spectrometry, we defined the repertoire of histone marks that are globally altered in quantity during reprogramming. Next, applying an in vivo CRISPR screening approach, we identified seven histone-modifying enzymes that alter the efficiency of hepatobiliary reprogramming. Among these, the histone methyltransferase and demethylase Nsd1 and Kdm2a were found to have reciprocal effects on H3K36 methylation that regulated the early and late stages of reprogramming, respectively. Although loss of Nsd1 and Kdm2a affected reprogramming efficiency, cells ultimately acquired the same transcriptomic states. These findings reveal that multiple chromatin regulators exert dynamic and complementary activities to achieve robust cell fate switching, serving as a model for the cell identity changes that occur in various forms of physiological metaplasia or reprogramming.
Collapse
Affiliation(s)
- Jonathan H Sussman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Hector W Cure
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Salina Yuan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Kenji Ito
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Irfan A Asangani
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Benjamin A Garcia
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Ben Z Stanger
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Takeshi Katsuda
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| |
Collapse
|
9
|
Lebegge E, Kancheva D, Van Craenenbroeck J, Ernst S, Bardet PMR, Caro AA, Kiss M, Jumapili NA, Barthelmess RM, Zivalj M, Assaf N, Ali L, Elkrim Y, Demuytere J, De Jonge J, Raes G, Hadadi E, Devoogdt N, Vincke C, Mohavedi K, Vereecke L, Ceelen W, Stijlemans B, Laoui D, Arnouk SM, Van Ginderachter JA. VSIG4-Expressing Macrophages Contribute to Antiparasitic and Antimetastatic Responses in the Peritoneal Cavity. Eur J Immunol 2025; 55:e202551804. [PMID: 40346775 PMCID: PMC12064866 DOI: 10.1002/eji.202551804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/12/2025]
Abstract
Large peritoneal macrophages (LPMs) play a role as gatekeepers of peritoneal homeostasis by providing a first line of defense against pathogens. A third of the LPMs express the surface receptor VSIG4, but it is unclear whether these cells differ from their VSIG4-negative counterparts and perform dedicated functions. We demonstrate that VSIG4+, but not VSIG4-, LPMs are in the majority derived from embryonal precursors, and their occurrence is largely independent of sex and microbiota but increases with age. Although their transcriptome and surface proteome are indistinguishable from VSIG4- LPMs at steady-state, VSIG4+ LPMs are superior in phagocytosing S. aureus bioparticles and colorectal carcinoma (CRC) cells. Anti-VSIG4 nanobody constructs that are ADCC-enabled allowed a selective elimination of the VSIG4+ LPM subset without affecting overall LPM abundance. This strategy uncovered a role for VSIG4+ LPMs in lowering the first peak of parasitemia in a Trypanosoma brucei brucei infection model and in reducing CRC outgrowth in the peritoneal cavity, a prime metastatic site in CRC patients. Altogether, our data uncover a protective role for VSIG4+ LPMs in infectious and oncological diseases in the peritoneal cavity.
Collapse
|
10
|
Muñoz Zamora A, Douglas A, Conway PB, Urrieta E, Moniz T, O'Leary JD, Marks L, Denny CA, Ortega-de San Luis C, Lynch L, Ryan TJ. Cold memories control whole-body thermoregulatory responses. Nature 2025; 641:942-951. [PMID: 40269165 PMCID: PMC12095059 DOI: 10.1038/s41586-025-08902-6] [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: 02/16/2024] [Accepted: 03/14/2025] [Indexed: 04/25/2025]
Abstract
Environmental thermal challenges trigger the brain to coordinate both autonomic and behavioural responses to maintain optimal body temperature1-4. It is unknown how temperature information is precisely stored and retrieved in the brain and how it is converted into a physiological response. Here we investigated whether memories could control whole-body metabolism by training mice to remember a thermal challenge. Mice were conditioned to associate a context with a specific temperature by combining thermoregulatory Pavlovian conditioning with engram-labelling technology, optogenetics and chemogenetics. We report that if mice are returned to an environment in which they previously experienced a 4 °C cold challenge, they increase their metabolic rates regardless of the actual environmental temperature. Furthermore, we show that mice have increased hypothalamic activity when they are exposed to the cold, and that a specific network emerges between the hippocampus and the hypothalamus during the recall of a cold memory. Both natural retrieval and artificial reactivation of cold-sensitive memory engrams in the hippocampus mimic the physiological responses that are seen during a cold challenge. These ensembles are necessary for cold-memory retrieval. These findings show that retrieval of a cold memory causes whole-body autonomic and behavioural responses that enable mice to maintain thermal homeostasis.
Collapse
Affiliation(s)
- Andrea Muñoz Zamora
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Aaron Douglas
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Paul B Conway
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Esteban Urrieta
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Taylor Moniz
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY, USA
| | - James D O'Leary
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Lydia Marks
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
- Allen Institute, Seattle, WA, USA
| | - Christine A Denny
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY, USA
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene (RFMH), New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Clara Ortega-de San Luis
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
- Department of Health Sciences, University of Jaén, Jaén, Spain
| | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland.
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland.
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia.
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Acebedo AR, Yamada G, Alcantara MC, Raga DD, Sato T, Nishinakamura R, Suzuki K. Sall1 regulates microtubule acetylation in mesenchymal cells during mouse urethral development. Cells Dev 2025:204027. [PMID: 40306366 DOI: 10.1016/j.cdev.2025.204027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 04/17/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Male embryonic external genitalia (eExG) undergo sexually dimorphic urethral development in response to androgen signaling (urethral masculinization). Whereas androgen is an essential masculinization factor for eExG, the specific molecular and cellular mechanisms are still unclear. Sall1 is a transcription factor that has been linked to the congenital disease Townes-Brocks syndrome, which includes anorectal and urogenital malformations. Currently, the functional role of Sall1 for normal urethral development is still unclear. In this study, we show that Sall1 is required to regulate proper microtubule acetylation to facilitate mesenchymal cell migration during urethral masculinization of mouse eExG. Mutant male mice with loss of function of mesenchymal Sall1 exhibited severe urethral defects, without prominent alteration of androgen signaling. Loss of Sall1 induced hyperacetylated microtubules in the eExG mesenchyme. Microtubule hyperacetylation resulted in defective fibrillar adhesions and fibronectin expression which impaired cell migration. Our findings reveal a novel mechanism of Sall1-regulated mesenchymal cell migration for urethral development. This mechanism for Sall1 may underlie the etiology of diseases such as Townes-Brocks syndrome.
Collapse
Affiliation(s)
- Alvin R Acebedo
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Institute of Biology, College of Science, University of the Philippines, Diliman, 1101 Quezon City, NCR, Philippines
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Department of Plastic and Reconstructive Surgery, Graduate School of Medicine Wakayama Medical University, Kimiidera, Wakayama 641-8509, Japan
| | - Mellissa C Alcantara
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Dennis D Raga
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Tetsuya Sato
- Biomedical Research Center, Faculty of Medicine, Saitama Medical University, Saitama 350-1241, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Kentaro Suzuki
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi 400-8510, Japan.
| |
Collapse
|
12
|
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
|
13
|
Luo YE, Abe-Teh Z, Alsaghir TY, Kuo LY, Yu F, Stoker BE, Appu AB, Zhou Y, Yue F, Kopinke D, Barton ER. Fibro-Adipogenic Progenitors require autocrine IGF-I in homeostatic and regenerating skeletal muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.11.648330. [PMID: 40291730 PMCID: PMC12027368 DOI: 10.1101/2025.04.11.648330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Fibro-Adipogenic Progenitors (FAPs) are mesenchymal stem cells that are vital for muscle homeostasis and regeneration but produce fibrosis and intramuscular fat under pathological conditions. Insulin-like Growth Factor-I (IGF-I) is a key regulator of muscle repair, satellite cell activity, macrophage polarization, and extracellular matrix (ECM) remodeling. We generated inducible FAP-specific Igf1 deficient (FID) mice to determine the necessity of FAP IGF-I. After BaCl 2 injury, FID mice exhibited impaired muscle regeneration, with fewer Pax7+ cells, increased macrophage accumulation, smaller fibers, reduced ECM, and depressed FAP proliferation. Following glycerol injury, FID muscles exhibited reduced adipocyte accumulation. Primary FAPs isolated from injured FID muscles had blunted growth, upregulation of immune-regulatory genes and downregulation of ECM and cell proliferation genes, with delayed responses to fibrogenic and to adipogenic media. FAP property alterations were already present in homeostatic muscle, indicated by scRNASeq, with decreased indices of protein translation and ECM production as well as increased markers of senescence, confirmed in vivo and in vitro . Overall, FAP IGF-I is a critical autocrine factor, with further paracrine consequences for muscle regenerative capacity.
Collapse
|
14
|
Tan SN, Hao J, Ge J, Yang Y, Liu L, Huang J, Lin M, Zhao X, Wang G, Yang Z, Ni L, Dong C. Regulatory T cells converted from Th1 cells in tumors suppress cancer immunity via CD39. J Exp Med 2025; 222:e20240445. [PMID: 39907686 PMCID: PMC11797014 DOI: 10.1084/jem.20240445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/17/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025] Open
Abstract
Regulatory T (Treg) cells are known to impede antitumor immunity, yet the regulatory mechanisms and functional roles of these cells remain poorly understood. In this study, through the characterization of multiple cancer models, we identified a substantial presence of peripherally induced Treg cells in the tumor microenvironment (TME). Depletion of these cells triggered antitumor responses and provided potent therapeutic effects by increasing functional CD8+ T cells. Fate-mapping and transfer experiments revealed that IFN-γ-expressing T helper (Th) 1 cells differentiated into Treg cells in response to TGF-β signaling in tumors. Pseudotime trajectory analysis further revealed the terminal differentiation of Th1-like Treg cells from Th1 cells in the TME. Tumor-resident Treg cells highly expressed T-bet, which was essential for their functions in the TME. Additionally, CD39 was highly expressed by T-bet+ Treg cells in both mouse and human tumors, and was necessary for Treg cell-mediated suppression of CD8+ T cell responses. Our study elucidated the developmental pathway of intratumoral Treg cells and highlighted novel strategies for targeting them in cancer patients.
Collapse
Affiliation(s)
- Sang-Nee Tan
- School of Medicine, Westlake University, Hangzhou, China
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Jing Hao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-affiliated Renji Hospital, Shanghai, China
| | - Jing Ge
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-affiliated Renji Hospital, Shanghai, China
| | - Yazheng Yang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Liguo Liu
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jia Huang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Meng Lin
- School of Medicine, Westlake University, Hangzhou, China
| | - Xiaohong Zhao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Genyu Wang
- School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiying Yang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Chen Dong
- School of Medicine, Westlake University, Hangzhou, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-affiliated Renji Hospital, Shanghai, China
| |
Collapse
|
15
|
Vicario R, Fragkogianni S, Pokrovskii M, Meyer C, Lopez-Rodrigo E, Hu Y, Ogishi M, Alberdi A, Baako A, Ay O, Plu I, Sazdovitch V, Heritier S, Cohen-Aubart F, Shor N, Miyara M, Nguyen-Khac F, Viale A, Idbaih A, Amoura Z, Rosenblum MK, Zhang H, Karnoub ER, Sashittal P, Jakatdar A, Iacobuzio-Donahue CA, Abdel-Wahab O, Tabar V, Socci ND, Elemento O, Diamond EL, Boisson B, Casanova JL, Seilhean D, Haroche J, Donadieu J, Geissmann F. Role of clonal inflammatory microglia in histiocytosis-associated neurodegeneration. Neuron 2025; 113:1065-1081.e13. [PMID: 40081365 DOI: 10.1016/j.neuron.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: 07/12/2023] [Revised: 10/28/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
Langerhans cell histiocytosis (LCH) and Erdheim-Chester disease (ECD) are clonal myeloid disorders associated with mitogen-activated protein (MAP)-kinase-activating mutations and an increased risk of neurodegeneration. We found microglial mutant clones in LCH and ECD patients, whether or not they presented with clinical symptoms of neurodegeneration, associated with microgliosis, astrocytosis, and neuronal loss, predominantly in the rhombencephalon gray nuclei. Neurological symptoms were associated with PU.1+ clone size (p = 0.0003) in patients with the longest evolution of the disease, indicating a phase of subclinical incipient neurodegeneration. Genetic barcoding analysis suggests that clones may originate from definitive or yolk sac hematopoiesis, depending on the patients. In a mouse model, disease topography was attributable to a local clonal proliferative advantage, and microglia depletion by a CSF1R-inhibitor limited neuronal loss and improved survival. These studies characterize a neurodegenerative disease associated with clonal proliferation of inflammatory microglia. The long preclinical stage represents a therapeutic window before irreversible neuronal depletion.
Collapse
Affiliation(s)
- Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stamatina Fragkogianni
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maria Pokrovskii
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carina Meyer
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Estibaliz Lopez-Rodrigo
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yang Hu
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell, New York, NY 10021, USA
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Araitz Alberdi
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ann Baako
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Oyku Ay
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Isabelle Plu
- Department of Neuropathology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Véronique Sazdovitch
- Department of Neuropathology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Sebastien Heritier
- French Langerhans cell histiocytosis registry, Department of Pediatric Hematology and Oncology, Trousseau Hospital, AP-HP, Paris, France
| | - Fleur Cohen-Aubart
- Department of Internal Medicine & Institut E3M, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Natalia Shor
- Department of Neuroradiology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Makoto Miyara
- Center for Immunology and Infectious Diseases (CIMI-PARIS), Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Florence Nguyen-Khac
- Department of Hematology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Agnes Viale
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, MSKCC, New York, NY 10065, USA
| | - Ahmed Idbaih
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié-Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, 75013 Paris, France
| | - Zahir Amoura
- Department of Neuroradiology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | | | - Haochen Zhang
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | | | - Palash Sashittal
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Akhil Jakatdar
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Christine A Iacobuzio-Donahue
- Department of Pathology, MSKCC, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Viviane Tabar
- Department of Neurosurgery, and Center for Stem Cell Biology, MSKCC, New York, NY, USA; Department of Neurology, MSKCC, New York, NY 10065, USA
| | - Nicholas D Socci
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, MSKCC, New York, NY 10065, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell, New York, NY 10021, USA
| | - Eli L Diamond
- Department of Neurosurgery, and Center for Stem Cell Biology, MSKCC, New York, NY, USA
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Danielle Seilhean
- Department of Neuropathology, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France
| | - Julien Haroche
- Department of Internal Medicine & Institut E3M, Pitié-Salpêtrière Hospital, APHP-Sorbonne Université, Paris, France.
| | - Jean Donadieu
- French Langerhans cell histiocytosis registry, Department of Pediatric Hematology and Oncology, Trousseau Hospital, AP-HP, Paris, France
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
16
|
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
|
17
|
Zhu W, Pan L, Cui X, Russo AC, Ray R, Pederson B, Wei X, Lin LL, Hafner H, Gregg B, Shrestha N, Liu C, Naji A, Arvan P, Sandoval DA, Lindberg I, Qi L, Reinert RB. SEL1L-HRD1 ER-Associated Degradation Facilitates Prohormone Convertase 2 Maturation and Glucagon Production in Islet α Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644437. [PMID: 40166183 PMCID: PMC11957139 DOI: 10.1101/2025.03.20.644437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Proteolytic cleavage of proglucagon by prohormone convertase 2 (PC2) is required for islet α cells to generate glucagon. However, the regulatory mechanisms underlying this process remain largely unclear. Here, we report that SEL1L-HRD1 endoplasmic reticulum (ER)-associated degradation (ERAD), a highly conserved protein quality control system responsible for clearing misfolded proteins from the ER, plays a key role in glucagon production by regulating turnover of the nascent proform of the PC2 enzyme (proPC2). Using a mouse model with SEL1L deletion in proglucagon-expressing cells, we observed a progressive decline in stimulated glucagon secretion and a reduction in pancreatic glucagon content. Mechanistically, we found that endogenous proPC2 is a substrate of SEL1L-HRD1 ERAD, and that degradation of misfolded proPC2 ensures the maturation of activation-competent proPC2 protein. These findings identify ERAD as a novel regulator of PC2 biology and an essential mechanism for maintaining α cell function.
Collapse
Affiliation(s)
- Wenzhen Zhu
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Linxiu Pan
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Present address: Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Xianwei Cui
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Anna Chiara Russo
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Rohit Ray
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Brent Pederson
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Xiaoqiong Wei
- Present address: Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Liangguang Leo Lin
- Present address: Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Hannah Hafner
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Brigid Gregg
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48105, USA
| | - Neha Shrestha
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Chengyang Liu
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ali Naji
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Darleen A. Sandoval
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pediatrics, Nutrition Section, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland-Baltimore, Baltimore, MD 21201, USA
| | - Ling Qi
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Present address: Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Rachel B. Reinert
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| |
Collapse
|
18
|
Hossain MK, Unger L, Larsen U, Altankhuyag A, Legøy TA, Paulo JA, Vethe H, Ghila L. Mapping the initial effects of carcinogen-induced oncogenic transformation in the mouse bladder. Exp Cell Res 2025; 446:114452. [PMID: 39988124 DOI: 10.1016/j.yexcr.2025.114452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
Characterizing the initial stages of oncogenic transformation allows the identification of tumor-promoting processes before the inherent clonal selection of the aggressive clones. Here, we used global proteomics, genetic cell tracing, and immunofluorescence to dynamically map the very early stages of cancer initiation in a mouse model of bladder cancer. We observed a very rapid and incremental proteome dysregulation, with changes in the energy metabolism, proliferation and immune signatures dominating the landscape. The changes in the lipid metabolism were immediate and defined by an increase fatty acid metabolism and lipid transport, followed by the activation of the immune landscape. Alongside the changes in the immune signature and lipid metabolism, we also mapped a clear increase in the cell cycle-related pathways and proliferation. Proliferation was mainly restricted to the basal epithelial layer rapidly leading to urothelium thickening, despite the progressive loss of the superficial layer. Moreover, we observed a tilt in the energy balance towards increased glucose metabolism, probably characterizing cells of the tumor microenvironment. All of the observed proteome signature changes were persistent, being retained and sometimes intensified or diversified along the timeline. The signatures observed in this pilot suggest these processes as potentially targetable drivers of the future neoplastic transformations in the bladder.
Collapse
Affiliation(s)
- Md Kaykobad Hossain
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Lucas Unger
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Ulrik Larsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | | | - Thomas Aga Legøy
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Heidrun Vethe
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Norway
| | - Luiza Ghila
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway.
| |
Collapse
|
19
|
Van den Berge K, Bakalar D, Chou HJ, Kunda D, Risso D, Street K, Purdom E, Dudoit S, Ngai J, Heavner W. A Latent Activated Olfactory Stem Cell State Revealed by Single-Cell Transcriptomic and Epigenomic Profiling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.26.564041. [PMID: 37961539 PMCID: PMC10634988 DOI: 10.1101/2023.10.26.564041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The olfactory epithelium is one of the few regions of the nervous system that sustains neurogenesis throughout life. Its experimental accessibility makes it especially tractable for studying molecular mechanisms that drive neural regeneration in response to injury. In this study, we used single-cell sequencing to identify the transcriptional cascades and epigenetic processes involved in determining olfactory epithelial stem cell fate during injury-induced regeneration. By combining gene expression and accessible chromatin profiles of individual lineage-traced olfactory stem cells, we identified transcriptional heterogeneity among activated stem cells at a stage when cell fates are being specified. We further identified a subset of resting cells that appears poised for activation, characterized by accessible chromatin around wound response and lineage-specific genes prior to their later expression in response to injury. Together these results provide evidence for a latent activated stem cell state, in which a subset of quiescent olfactory epithelial stem cells are epigenetically primed to support injury-induced regeneration.
Collapse
Affiliation(s)
- Koen Van den Berge
- Department of Statistics, University of California, Berkeley, CA
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Belgium
| | - Dana Bakalar
- Molecular Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Hsin-Jung Chou
- Department of Molecular and Cell Biology, University of California, Berkeley, CA
| | - Divya Kunda
- Molecular Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Davide Risso
- Department of Statistical Sciences, University of Padova, Padova, Italy
| | - Kelly Street
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Elizabeth Purdom
- Department of Statistics, University of California, Berkeley, CA
| | - Sandrine Dudoit
- Department of Statistics, University of California, Berkeley, CA
- Division of Biostatistics, School of Public Health, University of California, Berkeley, CA
| | - John Ngai
- Molecular Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Whitney Heavner
- Molecular Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| |
Collapse
|
20
|
Guo Y, Zhu M, Yu Z, Li Q, Chen Y, Ci L, Sun R, Shen R. Generation and characterization of a tamoxifen-inducible lineage tracing tool Cd2-P2A-CreERT2 knock-in mice. Front Immunol 2025; 16:1482070. [PMID: 40129982 PMCID: PMC11931051 DOI: 10.3389/fimmu.2025.1482070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Introduction The new targeted gene editing technologies, such as the CRISPR/Cas system, enable researchers to insert or delete genes at targeted loci efficiently. The Cre-loxp recombination system is widely used to activate or inactivate genes with high spatial and temporal specificity. Methods Using the CRISPR/Cas9 system, we inserted the CreERT2 transgene expression cassette into the Cd2 gene locus to generate conditional Cre-driver line Cd2-CreERT2 knock-in mice, which drove the expression of CreERT2 by the endogenous Cd2 promoter. By mating the Cd2-CreERT2 strain with a Rosa26-LSL-tdTomato reporter mouse strain which contains a tdTomato expression fragment blocked with a loxP-flanked STOP cassette (LSL) driven by a CAG promoter, a Cd2-CreERT2;Rosa26-LSL-tdTomato reporter strain was obtained to evaluate the expression pattern of CD2 in different cell types. Results After treatment with tamoxifen, the Cd2-CreERT2 knock-in mice were induced to perform efficient recombination at the loxP site following CreERT2 activation and cause the expression of tdTomato fluorescence. The tdTomato and CD2 were expressed in the T cells of peripheral blood, spleen and mesenteric lymph nodes, whereas detected in a low proportion in the B cells. While about 20% of cells labeled with tamoxifen-induced tdTomato were CD2+ monocytes in peripheral blood, 10% of dendritic cells were tdTomato+/CD2+ cells. Tamoxifen-independent expression of tdTomato occurred in approximately 3% of CD2+ macrophages, but in negligible (~0.5%) in CD2+ granulocytes. Discussion This work supplied a new transgenic mouse as a valuable tool for lineage tracing in CD2-expressing cells, for conditional mutant studies of immune modulatory effects in a time-dependent manner, and analysis of the potential therapeutic effect of CD2-targeting biologics.
Collapse
Affiliation(s)
- Yang Guo
- Model Organism R&D Department, Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Mengyan Zhu
- Model Organism R&D Department, Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Zhilan Yu
- Model Organism R&D Department, Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Qing Li
- Shanghai Engineering Research Center for Model Organizations, Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Yanjuan Chen
- Model Organism R&D Department, Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Lei Ci
- Shanghai Engineering Research Center for Model Organizations, Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Ruilin Sun
- Shanghai Engineering Research Center for Model Organizations, Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Ruling Shen
- Model Organism R&D Department, Shanghai Laboratory Animal Research Center, Shanghai, China
| |
Collapse
|
21
|
Higashiyama H, Kuroda S, Iwase A, Irie N, Kurihara H. On the Maxillofacial Development of Mice, Mus musculus. J Morphol 2025; 286:e70032. [PMID: 40022452 PMCID: PMC11871421 DOI: 10.1002/jmor.70032] [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: 11/14/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 03/03/2025]
Abstract
The maxillofacial region is one of the most complex areas in the vertebrate body plan. The homology of the upper jaw bones remain controversial, both between mammals and nonmammalian amniotes and among humans and other mammals, leading to various hypotheses on how this region evolved from ancestral amniotes to humans. As a key mammalian model, the mouse (Mus musculus) is vital for unraveling the evolution and development of the maxillofacial region experimentally. However, limited detailed morphological descriptions of murine cranial development hinder the extrapolation of findings to other species, including humans. Here, we describe the development of the murine face, including the nerves, skeletons, and vasculatures from the pharyngula (9.0 days post-coitum [dpc]) to the late fetal period (18.5 dpc) based on three-dimensional reconstructions of histological sections. The present results confirm that the morphology of the pharyngula stages and developmental process of chondrocranium of mice is highly conserved when compared to nonmammalian tetrapods and humans. We also propose that the Os incisivum, the rostralmost bone in the mammalian upper jaw, consists of septomaxillary and palatine components, supporting our previous hypothesis that the ancestral premaxilla was entirely lost in mammals. The present descriptive study of mice strengthen the anatomical correspondence between mouse and human faces and offers a solid framework for comparative craniofacial studies across vertebrates.
Collapse
Affiliation(s)
- Hiroki Higashiyama
- Research Center for Integrative Evolutionary Science (RCIES), The Graduate University for Advanced Studies, SOKENDAIHayamaKanagawaJapan
- Department of Physiological Chemistry and MetabolismGraduate School of Medicine, The University of TokyoTokyoBunkyo‐kuJapan
| | - Shunya Kuroda
- Institute for Frontier Science InitiativeKanazawa University, Kakuma‐machiKanazawaIshikawaJapan
| | - Akiyasu Iwase
- Department of Physiological Chemistry and MetabolismGraduate School of Medicine, The University of TokyoTokyoBunkyo‐kuJapan
- Isotope Science Center, The University of TokyoTokyoBunkyo‐kuJapan
| | - Naoki Irie
- Research Center for Integrative Evolutionary Science (RCIES), The Graduate University for Advanced Studies, SOKENDAIHayamaKanagawaJapan
| | - Hiroki Kurihara
- Department of Physiological Chemistry and MetabolismGraduate School of Medicine, The University of TokyoTokyoBunkyo‐kuJapan
- Isotope Science Center, The University of TokyoTokyoBunkyo‐kuJapan
- International Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoChuo‐kuJapan
| |
Collapse
|
22
|
Franceschini A, Jin M, Chen CW, Silvestri L, Mastrodonato A, Denny CA. Brain-wide immunolabeling and tissue clearing applications for engram research. Neurobiol Learn Mem 2025; 218:108032. [PMID: 39922482 DOI: 10.1016/j.nlm.2025.108032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
In recent years, there has been significant progress in memory research, driven by genetic and imaging technological advances that have given unprecedented access to individual memory traces or engrams. Although Karl Lashley argued since the 1930s that an engram is not confined to a particular area but rather distributed across the entire brain, most current studies have focused exclusively on a single or few brain regions. However, this compartmentalized approach overlooks the interactions between multiple brain regions, limiting our understanding of engram mechanisms. More recently, several studies have begun to investigate engrams across the brain, but research is still limited by a lack of standardized techniques capable of reconstructing multiple ensembles at single-cell resolution across the entire brain. In this review, we guide researchers through the latest technological advancements and discoveries in immediate early gene (IEG) techniques, tissue clearing methods, microscope modalities, and automated large-scale analysis. These innovations could propel the field forward in building brain-wide engram maps of normal and disease states, thus, providing unprecedented new insights. Ultimately, this review aims to bridge the gap between research focused on single brain regions and the need for a comprehensive understanding of whole-brain engrams, revealing new approaches for exploring the neuronal mechanisms underlying engrams.
Collapse
Affiliation(s)
- Alessandra Franceschini
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, 50019 Italy
| | - Michelle Jin
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; Neurobiology and Behavior (NB&B) Graduate Program, Columbia University, New York, NY 10027, USA
| | - Claire W Chen
- Cellular, Molecular, and Biomedical Sciences Graduate Program, Columbia University, New York, NY 10027, USA
| | - Ludovico Silvestri
- European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, 50019 Italy; Department of Physics and Astronomy, University of Florence, Sesto Fiorentino 50019, Italy
| | - Alessia Mastrodonato
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY 10032, USA.
| | - Christine Ann Denny
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY 10032, USA.
| |
Collapse
|
23
|
Aoki H, Tomita H, Hara A, Kunisada T. Conditional heterozygous loss of kit receptor tyrosine kinase in neural crest cell lineage is associated with midline cleft lip and bifid nose deformity. J Oral Biosci 2025; 67:100572. [PMID: 39426597 DOI: 10.1016/j.job.2024.10.004] [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: 07/18/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVES The receptor tyrosine kinase Kit is expressed in cells derived from the trunk neural crest (NC), such as melanocytes; however, its role in cranial NC cell development is not fully understood. METHODS We investigated the effects of the heterozygous loss of Kit in NC cells during embryonic development by mating Kit2lox/+ mice with Wnt1-Cre mice to produce Wnt1-Cre; Kit2lox/+ embryos. In addition, Wnt1-Cre mice were mated with Rosa26R-yellow fluorescent protein (YFP) mice to visualize the tissue regions expressing Cre recombinase. Histological studies of the craniofacial regions of these mice were performed using samples from embryonic day (E) 12.5 and postnatal day (P) 1. Cellular apoptosis and proliferation were both analyzed through the immunostaining of tissue sections collected on E13.5 and E14.5 using anti-cleaved caspase 3 (CC3) to detect apoptosis and anti-Ki67 to detect proliferation. Cells from YFP-positive tissue regions of the facial areas of Wnt1-Cre; Kit+/+; Rosa26R-YFP embryos and Wnt1-Cre; Kit2lox/+; Rosa26R-YFP embryos collected on E12.5 and E15.5 were cultured and evaluated for cell proliferation. RESULTS Compared with control littermates, Wnt1-Cre; Kit2lox/+ embryos exhibited midline cleft lip and bifid nose deformities. Substantial early (P1) postnatal lethality was observed in Wnt1-Cre; Kit2lox/+ mice, with none surviving to 3 weeks of age. YFP-positive cells from the maxillary regions of Wnt1-Cre; Kit2lox/+; Rosa26R-YFP embryos exhibited defective cell growth and self-renewal in vitro. CONCLUSION Conditional heterozygous loss of Kit in Wnt1-Cre; Kit2lox/+ embryos is associated with craniofacial dysplasia and exhibit defective NC development in vitro and in vivo.
Collapse
Affiliation(s)
- Hitomi Aoki
- Department of Stem Cell and Regenerative Medicine, Gifu University Graduate School of Medicine, 1-1, Yanagido, Gifu, 501-1194, Japan.
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1, Yanagido, Gifu, 501-1194, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1, Yanagido, Gifu, 501-1194, Japan
| | - Takahiro Kunisada
- Department of Stem Cell and Regenerative Medicine, Gifu University Graduate School of Medicine, 1-1, Yanagido, Gifu, 501-1194, Japan
| |
Collapse
|
24
|
Rodrigues Sousa E, de Brot S, Zoni E, Zeinali S, Brunello A, Scarpa M, De Menna M, La Manna F, Abey Alexander A, Klima I, Freeman DW, Gates BL, Cristaldi DA, Guenat OT, Kruithof BPT, Spike BT, Chouvardas P, Kruithof-de Julio M. CRIPTO's multifaceted role in driving aggressive prostate cancer unveiled by in vivo, organoid, and patient data. Oncogene 2025; 44:462-475. [PMID: 39592836 PMCID: PMC11810784 DOI: 10.1038/s41388-024-03230-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024]
Abstract
CRIPTO (or CR-1 or TDGF1) is a protein that plays an active role in tumor initiation and progression. We have confirmed that increased expression of CRIPTO is associated with clinical and prostate-specific antigen (PSA) progression in human prostate tissues. Our approach involved gaining insight into the role of CRIPTO signaling in castration-resistant Nkx3.1-expressing cells (CARNs), targets for oncogenic transformation in prostate cancer (PCa), by integrating the existing Criptoflox/flox into CARNs model. The most aggressive stage was modeled using an inducible Cre under control of the Nkx3.1 promoter conferring Nkx3.1 inactivation and driving Pten inactivation, oncogenic Kras activation, and lineage tracing with yellow fluorescence protein (EYFP) upon induction. Our findings provide evidence that selective depletion of Cripto in epithelial cells in vivo reduced the invasive phenotype, particularly in more advanced tumor stages. Moreover, in vitro experiments with Cripto overexpression demonstrated alterations in the physical features of organoids, which correlated with increased tumorigenic activity. Transcriptomic analyses revealed a unique CRIPTO/MYC co-activation signature associated with PSA progression in a human PCa cohort. Taken together, our data highlights a role for CRIPTO in tumor invasiveness and progression, which carries implications for biomarkers and targeted therapies.
Collapse
Affiliation(s)
- Elisa Rodrigues Sousa
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Simone de Brot
- COMPATH, Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Eugenio Zoni
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Soheila Zeinali
- Organs-on-chip Technologies Laboratory, ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Andrea Brunello
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Mario Scarpa
- Department for BioMedical Research, Translational Organoid Research, University of Bern, Bern, Switzerland
| | - Marta De Menna
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department for BioMedical Research, Translational Organoid Research, University of Bern, Bern, Switzerland
| | - Federico La Manna
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Allen Abey Alexander
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Irena Klima
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - David W Freeman
- Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, USA
| | - Brooke L Gates
- Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, USA
| | | | - Olivier T Guenat
- Organs-on-chip Technologies Laboratory, ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Boudewijn P T Kruithof
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Benjamin T Spike
- Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, USA
| | - Panagiotis Chouvardas
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland.
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Department for BioMedical Research, Translational Organoid Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
25
|
Yampolsky M, Bachelet I, Fuchs Y. Wound localization and housing conditions dictate repair dynamics and scar formation. Lab Anim (NY) 2025; 54:68-73. [PMID: 40011792 DOI: 10.1038/s41684-025-01520-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 01/28/2025] [Indexed: 02/28/2025]
Abstract
Wound healing is a highly orchestrated process involving diverse cells and molecular interplays. Although wound healing assays are commonly used in the field of tissue repair, these experiments exhibit high variability due to their multifactorial nature, with many design factors remaining understudied. Here we report that precise localization of the wound site as well as the housing conditions have a pivotal role in dictating the healing dynamics in mice. We explore these key factors and highlight overlooked aspects of the repair process.
Collapse
|
26
|
Lehtonen H, Jokela H, Hofmann J, Tola L, Mehmood A, Ginhoux F, Becher B, Greter M, Yegutkin GG, Salmi M, Gerke H, Rantakari P. Early precursor-derived pituitary gland tissue-resident macrophages play a pivotal role in modulating hormonal balance. Cell Rep 2025; 44:115227. [PMID: 39841599 DOI: 10.1016/j.celrep.2024.115227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/26/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025] Open
Abstract
The pituitary gland is the central endocrine regulatory organ producing and releasing hormones that coordinate major body functions. The physical location of the pituitary gland at the base of the brain, though outside the protective blood-brain barrier, leads to an unexplored special immune environment. Using single-cell transcriptomics, fate mapping, and imaging, we characterize pituitary-resident macrophages (pitMØs), revealing their heterogeneity and spatial specialization. Microglia-like macrophages (ml-MACs) are enriched in the posterior pituitary, while other pitMØs in the anterior pituitary exhibit close interactions with hormone-secreting cells. Importantly, all pitMØs originate from early yolk sac progenitors and maintain themselves through self-renewal, independent of bone marrow-derived monocytes. Macrophage depletion experiments unveil the role of macrophages in regulating intrapituitary hormonal balance through extracellular ATP-mediated intercellular signaling. Altogether, these findings provide information on pituitary gland macrophages and advance our understanding of immune-endocrine system crosstalk.
Collapse
Affiliation(s)
- Henna Lehtonen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Heli Jokela
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Julian Hofmann
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Lauriina Tola
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Arfa Mehmood
- Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore; INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France; Translational Immunology Institute, SingHealth Duke-NUS, Singapore 169856, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Gennady G Yegutkin
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland; MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | - Marko Salmi
- Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland; MediCity Research Laboratory, University of Turku, 20520 Turku, Finland
| | - Heidi Gerke
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Pia Rantakari
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland.
| |
Collapse
|
27
|
Amo-Shiinoki K, Tanabe K, Nishimura W, Hatanaka M, Kondo M, Kagawa S, Zou M, Morikawa S, Sato Y, Komatsu M, Mizukami H, Nishida N, Asahara SI, Masutani H, Tanizawa Y. β cell dedifferentiation, the underlying mechanism of diabetes in Wolfram syndrome. Sci Transl Med 2025; 17:eadp2332. [PMID: 39970233 DOI: 10.1126/scitranslmed.adp2332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/06/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025]
Abstract
Insulin-dependent diabetes in patients with Wolfram syndrome (WS; OMIM 222300) has been linked to endoplasmic reticulum (ER) stress caused by WFS1 gene mutations. However, the pathological process of ER stress-associated β cell failure remains to be fully elucidated. Our results indicate loss of β cell lineage and subsequent dedifferentiation as the mechanisms underlying functional and mass deficits in WS. An immunohistochemical analysis of human pancreatic sections from deceased individuals with WS revealed a near-complete loss of β cells and subsequent decrease in α cells, suggesting loss of endocrine function. Wfs1-deficient mice displayed dysfunction, gradual loss, and dedifferentiation of β cells, leading to permanent hyperglycemia. Impairment of the β cell lineage was observed after weaning, leading to the mixed phenotype of insulin- and glucagon-producing cells in a subset of the lineage-traced β cells. Islets of Wfs1-deficient mice increased the number of dedifferentiated cells that maintained general endocrine features but were no longer reactive with antisera against pancreatic hormones. Mechanistically, Wfs1-null islets had a lower adenosine triphosphate content and impaired oxidative glycolysis, although mitochondrial oxidative function was maintained. The functional and metabolic alterations of WS β cells were recovered by deletion of thioredoxin-interacting protein (Txnip), an ER stress-induced protein up-regulated in Wfs1 deficiency. Txnip deletion preserved functional β cells and prevented diabetes progression in Wfs1-deficient mice. Together, this study deciphered pathological mechanisms of β cell dedifferentiation in β cell failure and has implications for Txnip inhibition in WS therapy.
Collapse
Affiliation(s)
- Kikuko Amo-Shiinoki
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
- Department of Diabetes Research, Yamaguchi University School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Katsuya Tanabe
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Wataru Nishimura
- Department of Molecular Biology, International University of Health and Welfare School of Medicine, Narita, Chiba 286-8686, Japan
- Department of Anatomy, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Masayuki Hatanaka
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Manabu Kondo
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Syota Kagawa
- Department of Natural Products Chemistry, Daiichi University of Pharmacy, Fukuoka, Fukuoka 815-8511, Japan
| | - Meng Zou
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Shuntaro Morikawa
- Department of Pediatrics, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Yoshihiko Sato
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Nagano 390-8621, Japan
- Department of Internal Medicine, Matsumoto City Hospital, Matsumoto, Nagano 390-1401, Japan
| | - Mitsuhisa Komatsu
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Nagano 390-8621, Japan
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Biomedical Research Center, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Naoki Nishida
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Shun-Ichiro Asahara
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Hiroshi Masutani
- Department of Clinical Laboratory Sciences, Faculty of Health Care, Tenri University, Tenri, Nara 632-0018, Japan
| | - Yukio Tanizawa
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
28
|
Jin M, Ogundare SO, Lanio M, Sorid S, Whye AR, Santos SL, Franceschini A, Denny CA. A SMARTTR workflow for multi-ensemble atlas mapping and brain-wide network analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.12.603299. [PMID: 39071434 PMCID: PMC11275872 DOI: 10.1101/2024.07.12.603299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
In the last decade, activity-dependent strategies for labelling multiple immediate early gene (IEG) ensembles in mice have generated unprecedented insight into the mechanisms of memory encoding, storage, and retrieval. However, few strategies exist for brain-wide mapping of multiple ensembles, including their overlapping population, and none incorporate capabilities for downstream network analysis. Here, we introduce a scalable workflow to analyze traditionally coronally-sectioned datasets produced by activity-dependent tagging systems. Intrinsic to this pipeline is simple multi-ensemble atlas registration and statistical testing in R (SMARTTR), an R package which wraps mapping capabilities with functions for statistical analysis and network visualization, and support for import of external datasets. We demonstrate the versatility of SMARTTR by mapping the ensembles underlying the acquisition and expression of learned helplessness (LH), a robust stress model. Applying network analysis, we find that exposure to inescapable shock (IS), compared to context training (CT), results in decreased centrality of regions engaged in spatial and contextual processing and higher influence of regions involved in somatosensory and affective processing. During LH expression, the substantia nigra emerges as a highly influential region which shows a functional reversal following IS, indicating a possible regulatory function of motor activity during helplessness. We also report that IS results in a robust decrease in reactivation activity across a number of cortical, hippocampal, and amygdalar regions, indicating suppression of ensemble reactivation may be a neurobiological signature of LH. These results highlight the emergent insights uniquely garnered by applying our analysis approach to multiple ensemble datasets and demonstrate the strength of our workflow as a hypothesis-generating toolkit.
Collapse
Affiliation(s)
- Michelle Jin
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
- Neurobiology and Behavior (NB&B) Graduate Program, Columbia University, New York, NY, 10027, USA
| | - Simon O. Ogundare
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
- Columbia College, New York, NY, 10027, USA
| | - Marcos Lanio
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
- Adult Neurology Residency Program, Stony Brook Medicine, Stony Brook, NY, 11794, USA
| | | | - Alicia R. Whye
- Columbia College, New York, NY, 10027, USA
- Tri-Institutional MD-PhD Program, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Sofia Leal Santos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
| | - Alessandra Franceschini
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
- European Laboratory for Non-linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Christine. A. Denny
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| |
Collapse
|
29
|
Jacques K, Coles BLK, van der Kooy D. Pancreatic stem cells originate during the pancreatic progenitor developmental stage. Front Cell Dev Biol 2025; 13:1521411. [PMID: 40040790 PMCID: PMC11876382 DOI: 10.3389/fcell.2025.1521411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Previously isolated adult pancreatic precursors called pancreatic multipotent progenitors (which make both pancreatic endocrine and exocrine cell types) originate from the Pancreatic Duodenal Homeobox 1 (PDX1) pancreatic developmental lineage. The embryonic time point at which adult pancreatic multipotent progenitor cells emerge has not been established. We have employed the use of two models: a human embryonic stem cell (hESC) to beta-cell cytokine-induced differentiation protocol and a mouse lineage tracing model during early development to isolate clonal pancreatic spheres. The results show that insulin-positive clonal spheres can be isolated as early as the pancreatic endoderm stage as well as the pancreatic progenitor stage during the hESC to beta-cell lineage differentiation model and that they can be isolated only as early as the pancreatic progenitor stage during mouse embryogenesis. Further, pancreatic clonal sphere-forming cells isolated from the pancreatic progenitor stage in embryonic mice display multipotentiality, and those isolated at a later gestational age demonstrate self-renewal ability. These findings suggest that pancreatic precursors isolated from mouse embryonic time points have stem cell properties and that the pancreatic progenitor stage in hESC development may be the optimal time to capture and expand these stem cells and make large numbers of beta cells.
Collapse
Affiliation(s)
- Krystal Jacques
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Brenda L. K. Coles
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Derek van der Kooy
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
30
|
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
|
31
|
Michalopulos MG, Liu Y, Raju DR, Lafin JT, Ma Y, Gaur D, Khadka S, Xing C, McMahon AP, Carroll TJ, Drake KA. Defects in nephrogenesis result in an expansion of the Foxd1+ stromal progenitor population. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637031. [PMID: 39990396 PMCID: PMC11844377 DOI: 10.1101/2025.02.10.637031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Reciprocal signaling interactions coordinate multiple aspects of kidney development. While signals from the stroma have been shown to regulate nephron progenitor cell (NPC) differentiation, much less is known about regulation of the stromal progenitor population. Here, we demonstrate that disruption of the NPC lineage via loss of Wt1 (i.e., Six2cre;Wt1 c/c ) results in an expansion of Foxd1+ stromal progenitor cells. Analyses of the developing stroma in two additional models, including Wnt4-null mutants (which fail to form nephron structures similar to Six2cre;Wt1 c/c kidneys) and NPC ablation via diphtheria toxin (i.e., Six2cre;RosaDTA c/+ ), both phenocopy Six2cre;Wt1 c/c mutants, thus further confirming that defects in the NPC lineage result in abnormal development of the stromal progenitor population. Furthermore, we identify a subcluster of the Foxd1+ stroma that appears expanded in the three mutant mouse models and conserved in human fetal kidneys. Overall, the findings from this study suggest that loss of differentiating nephron structures may result in possible over proliferation of the stromal progenitor population and/or a block in stromal differentiation and further highlight how crosstalk amongst the progenitor cell lineages coordinates multiple aspects of kidney development.
Collapse
Affiliation(s)
- Michael G Michalopulos
- Department of Pediatrics, Division of Pediatric Nephrology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Yan Liu
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dinesh Ravindra Raju
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John T Lafin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yanru Ma
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dhruv Gaur
- Midwestern University, Chicago College of Osteopathic Medicine, Chicago, IL, USA
| | - Sadiksha Khadka
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- Department of Bioinformatics and O'Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Thomas J Carroll
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Keri A Drake
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
32
|
Grootaert MOJ, Pasut A, Raman J, Simmonds SJ, Callewaert B, Col Ü, Dewerchin M, Carmeliet P, Heymans S, Jones EAV. Mural cell dysfunction contributes to diastolic heart failure by promoting endothelial dysfunction and vessel remodelling. Cardiovasc Diabetol 2025; 24:62. [PMID: 39920783 PMCID: PMC11806843 DOI: 10.1186/s12933-025-02623-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a complex cardiovascular disease associated with metabolic comorbidities. Microvascular dysfunction has been proposed to drive HFpEF, likely via endothelial cell (EC) dysfunction, yet the role of the mural cells herein has never been explored. METHODS We used the diabetic db/db mouse given 1% salt as a new model of HFpEF and crossed then with PDGFRβtg/tg-CreERT2-EYFPtg/tg mice to label the mural cells. We combined single-cell RNA sequencing, NichetNet analysis and histology to determine the role of mural cell dysfunction in HFpEF. RESULTS Db/db mice given 1% salt for 8 weeks developed diastolic dysfunction preceded by capillary density loss, pericyte loss and vessel regression. At 4 weeks of salt, hearts of db/db mice already showed EC dysfunction associated with an anti-angiogenic signature, and an increase in pericyte-EC intracellular space. Db/db + salt hearts were further characterised by increased ACTA2 expression, arteriole wall thickening and vessel enlargement. NicheNet analysis on the single cell transcriptomic data revealed little signalling from the ECs to the mural cells; instead, mural cells signalled strongly to ECs. Mechanistically, pericyte dysfunction induces an EC growth arrest via TNFα-dependent paracrine signalling and downstream signalling through STAT1. CONCLUSION Mural cell dysfunction contributes to HFpEF by inducing coronary vessel remodelling, at least in part by reducing EC proliferation and inducing EC inflammation through TNFα-dependent paracrine signalling.
Collapse
MESH Headings
- Animals
- Pericytes/metabolism
- Pericytes/pathology
- Disease Models, Animal
- Vascular Remodeling
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Heart Failure, Diastolic/physiopathology
- Heart Failure, Diastolic/metabolism
- Heart Failure, Diastolic/pathology
- Heart Failure, Diastolic/genetics
- Heart Failure, Diastolic/etiology
- Coronary Vessels/metabolism
- Coronary Vessels/physiopathology
- Coronary Vessels/pathology
- Signal Transduction
- Ventricular Function, Left
- Male
- Mice, Transgenic
- Mice, Inbred C57BL
- Stroke Volume
- Mice
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Paracrine Communication
- Actins
Collapse
Affiliation(s)
- Mandy O J Grootaert
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium.
- Endocrinology, Diabetes and Nutrition, UCLouvain, Avenue Hippocrate, box 55, Brussels, 1200, Belgium.
| | - Alessandra Pasut
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology (CCB), KU Leuven, Leuven, Belgium
| | - Jana Raman
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Steven J Simmonds
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Bram Callewaert
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Ümare Col
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology (CCB), KU Leuven, Leuven, Belgium
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology (CCB), KU Leuven, Leuven, Belgium
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Stephane Heymans
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
| | - Elizabeth A V Jones
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium.
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands.
| |
Collapse
|
33
|
Fimiani C, Pereira JA, Gerber J, Berg I, DeGeer J, Bachofner S, Fischer JS, Kauffmann M, Suter U. The E3 ubiquitin ligase Nedd4 fosters developmental myelination in the mouse central and peripheral nervous system. Glia 2025; 73:422-444. [PMID: 39511974 DOI: 10.1002/glia.24642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024]
Abstract
Ubiquitination is a major post-translational regulatory mechanism that tunes numerous aspects of ubiquitinated target proteins, including localization, stability, and function. During differentiation and myelination, Oligodendrocytes (OLs) in the central nervous system and Schwann cells (SCs) in the peripheral nervous system undergo major cellular changes, including the tightly controlled production of large and accurate amounts of proteins and lipids. Such processes have been implied to depend on regulatory aspects of ubiquitination, with E3 ubiquitin ligases being generally involved in the selection of specific ubiquitination substrates by ligating ubiquitin to targets and granting target selectivity. In this study, we have used multiple transgenic mouse lines to investigate the functional impact of the E3 ubiquitin ligase Nedd4 in the OL- and SC-lineages. Our findings in the developing spinal cord indicate that Nedd4 is required for the correct accumulation of differentiated OLs and ensures proper myelination, supporting and further expanding previously suggested conceptual models. In sciatic nerves, we found that Nedd4 is required for timely radial sorting of axons by SCs as a pre-requirement for correct onset of myelination. Moreover, Nedd4 ensures correct myelin thickness in both SCs and spinal cord OLs.
Collapse
Affiliation(s)
- Cristina Fimiani
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jorge A Pereira
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Joanne Gerber
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Ingrid Berg
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jonathan DeGeer
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Sven Bachofner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jonas S Fischer
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Manuel Kauffmann
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Ueli Suter
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Garcia-Gonzalez I, Gambera S, Rocha SF, Regano A, Garcia-Ortega L, Lytvyn M, Diago-Domingo L, Sanchez-Muñoz MS, Garcia-Cabero A, Zagorac I, Luo W, De Andrés-Laguillo M, Fernández-Chacón M, Casquero-Garcia V, Lunella FF, Torroja C, Sánchez-Cabo F, Benedito R. iFlpMosaics enable the multispectral barcoding and high-throughput comparative analysis of mutant and wild-type cells. Nat Methods 2025; 22:323-334. [PMID: 39672980 PMCID: PMC11810794 DOI: 10.1038/s41592-024-02534-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: 04/05/2024] [Accepted: 10/15/2024] [Indexed: 12/15/2024]
Abstract
To understand gene function, it is necessary to compare cells carrying the mutated target gene with normal cells. In most biomedical studies, the cells being compared are in different mutant and control animals and, therefore, do not experience the same epigenetic changes and tissue microenvironment. The experimental induction of genetic mosaics is essential to determine a gene cell-autonomous function and to model the etiology of diseases caused by somatic mutations. Current technologies used to induce genetic mosaics in mice lack either accuracy, throughput or barcoding diversity. Here we present the iFlpMosaics toolkit comprising a large set of new genetic tools and mouse lines that enable recombinase-dependent ratiometric induction and single-cell clonal tracking of multiple fluorescently labeled wild-type and Cre-mutant cells within the same time window and tissue microenvironment. The labeled cells can be profiled by multispectral imaging or by fluorescence-activated flow cytometry and single-cell RNA sequencing. iFlpMosaics facilitate the induction and analysis of genetic mosaics in any quiescent or progenitor cell, and for any given single or combination of floxed genes, thus enabling a more accurate understanding of how induced genetic mutations affect the biology of single cells during tissue development, homeostasis and disease.
Collapse
Affiliation(s)
- Irene Garcia-Gonzalez
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Stefano Gambera
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Susana F Rocha
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alvaro Regano
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Lourdes Garcia-Ortega
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Mariya Lytvyn
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Luis Diago-Domingo
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Maria S Sanchez-Muñoz
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Aroa Garcia-Cabero
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ivana Zagorac
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Wen Luo
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Macarena De Andrés-Laguillo
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Macarena Fernández-Chacón
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Verónica Casquero-Garcia
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Federica Francesca Lunella
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Carlos Torroja
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Rui Benedito
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
35
|
Wang H, Zhao B, Huang L, Zhu X, Li N, Huang C, Han Z, Ouyang K. Conditional deletion of IP 3R1 by Islet1-Cre in mice reveals a critical role of IP 3R1 in interstitial cells of Cajal in regulating GI motility. J Gastroenterol 2025; 60:152-165. [PMID: 39476178 DOI: 10.1007/s00535-024-02164-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/16/2024] [Indexed: 02/05/2025]
Abstract
BACKGROUND AND AIMS Inositol 1,4,5-trisphosphate receptor type 1 (IP3R1) has been proposed to play a physiological role in regulating gastrointestinal (GI) motility, but the underlying cell-dependent mechanism remains unclear. Here, we utilized cell-specific IP3R1 deletion strategies to address this question in mice. METHODS Conditional IP3R1 knockout mice using Wnt1-Cre, Islet1-Cre mice, and smMHC-CreEGFP were generated. Cell lineage tracing was performed to determine where gene deletion occurred in the GI tract. Whole-gut transit assay and isometric tension recording were used to assess GI function in vivo and in vitro. RESULTS In the mouse GI tract, Islet1-Cre targeted smooth muscle cells (SMCs) and interstitial cells of Cajal (ICCs), but not enteric neurons. IP3R1 deletion by Islet1-Cre (isR1KO) caused a phenotype of intestinal pseudo-obstruction (IPO), evidenced by prolonged whole-gut transit time, enlarged GI tract, abdominal distention, and early lethality. IP3R1 deletion by Islet1-Cre not only reduced the frequency of spontaneous contractions but also decreased the contractile responses to the muscarinic agonist carbachol (CCh) and electrical field stimulation (EFS) in colonic circular muscles. By contrast, smMHC-CreEGFP only targeted SMCs in the mouse GI tract. Although IP3R1 deletion by smMHC-CreEGFP (smR1KO) also reduced the contractile responses to CCh and EFS in colonic circular muscles, the frequency of spontaneous contractions was less affected, and neither global GI abnormalities nor early lethality was found in smR1KO mice. CONCLUSIONS IP3R1 deletion in both ICCs and SMCs but not in SMCs alone causes an IPO phenotype, suggesting that IP3R1 in ICCs plays an essential role in regulating GI motility in vivo.
Collapse
Affiliation(s)
- Hong Wang
- Central Laboratory, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, 518036, China
| | - Beili Zhao
- Central Laboratory, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, 518036, China
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Xiangbin Zhu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Na Li
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Can Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China.
| | - Kunfu Ouyang
- Central Laboratory, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, 518036, China.
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China.
| |
Collapse
|
36
|
Xypolita ME, Goolam M, Bikoff EK, Robertson EJ, Mould AW. The zinc-finger transcription factor Blimp1/Prdm1 is required for uterine remodelling and repair in the mouse. Nat Commun 2025; 16:1220. [PMID: 39890816 PMCID: PMC11785775 DOI: 10.1038/s41467-025-56511-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/17/2025] [Indexed: 02/03/2025] Open
Abstract
The zinc finger transcription factor Blimp1/PRDM1 regulates gene expression in diverse cell types. Its activity controls the maternal decidual response at early post-implantation stages of development. The present experiments demonstrate surprisingly that Blimp1 activity in the uterus is required for tissue remodelling at sites of embryonic failure. Moreover Blimp1 mutant females are refractory to RU486 induced decidual shedding. RNA-seq together with immunostaining experiments strongly suggest that the failure to up-regulate expression of the matrix metalloprotease Mmp10 in combination with insufficient suppression of BMP signalling, likely explain Blimp1-dependent phenotypic changes. In the post-partum uterus Blimp1 together with Mmp10 are highly upregulated at sites of tissue repair following placental detachment. Conditional Blimp1 removal significantly impairs the re-epithelization process and severely impacts involution of the endometrium and luminal epithelium. Overall these results identify Blimp1 as a master regulator of uterine tissue remodelling and repair.
Collapse
Affiliation(s)
- Maria-Eleni Xypolita
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Mubeen Goolam
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- Department of Human Biology and Neuroscience Institute, University of Cape Town, Cape Town, 7925, South Africa
| | - Elizabeth K Bikoff
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Elizabeth J Robertson
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
| | - Arne W Mould
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| |
Collapse
|
37
|
Fang LP, Lin CH, Medlej Y, Zhao R, Chang HF, Guo Q, Wu Z, Su Y, Zhao N, Gobbo D, Wyatt A, Wahl V, Fiore F, Tu SM, Boehm U, Huang W, Bian S, Agarwal A, Lauterbach MA, Yi C, Niu J, Scheller A, Kirchhoff F, Bai X. Oligodendrocyte precursor cells facilitate neuronal lysosome release. Nat Commun 2025; 16:1175. [PMID: 39885146 PMCID: PMC11782495 DOI: 10.1038/s41467-025-56484-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/17/2025] [Indexed: 02/01/2025] Open
Abstract
Oligodendrocyte precursor cells (OPCs) shape brain function through many non-canonical regulatory mechanisms beyond myelination. Here we show that OPCs form contacts with their processes on neuronal somata in a neuronal activity-dependent manner. These contacts facilitate exocytosis of neuronal lysosomes. A reduction in the number or branching of OPCs reduces these contacts, which is associated with lysosome accumulation and altered metabolism in neurons and more senescent neurons with age. A similar reduction in OPC branching and neuronal lysosome accumulation is seen in an early-stage mouse model of Alzheimer's disease. Our findings have implications for the prevention of age-related pathologies and the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Li-Pao Fang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 211198, Nanjing, China
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
| | - Ching-Hsin Lin
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Cellular Neurophysiology, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Yasser Medlej
- Molecular Imaging, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Renping Zhao
- Biophysics, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Hsin-Fang Chang
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Cellular Neurophysiology, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Qilin Guo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
| | - Zhonghao Wu
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, 518107, Shenzhen, China
| | - Yixun Su
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, 518107, Shenzhen, China
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, 400038, Chongqing, China
| | - Na Zhao
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
- Institute of Anatomy and Cell Biology, University of Saarland, 66421, Homburg, Germany
| | - Davide Gobbo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
| | - Amanda Wyatt
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Vanessa Wahl
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Frederic Fiore
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Szu-Min Tu
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Cellular Neurophysiology, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Ulrich Boehm
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Wenhui Huang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
| | - Shan Bian
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Amit Agarwal
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | | | - Chenju Yi
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, 400038, Chongqing, China
| | - Jianqin Niu
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, 518107, Shenzhen, China
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany.
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany.
| | - Xianshu Bai
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany.
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 211198, Nanjing, China.
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany.
| |
Collapse
|
38
|
Lozovska A, Casaca A, Novoa A, Kuo YY, Jurberg AD, Martins GG, Hadjantonakis AK, Mallo M. Tgfbr1 regulates lateral plate mesoderm and endoderm reorganization during the trunk to tail transition. eLife 2025; 13:RP94290. [PMID: 39874182 PMCID: PMC11774515 DOI: 10.7554/elife.94290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
During the trunk to tail transition the mammalian embryo builds the outlets for the intestinal and urogenital tracts, lays down the primordia for the hindlimb and external genitalia, and switches from the epiblast/primitive streak (PS) to the tail bud as the driver of axial extension. Genetic and molecular data indicate that Tgfbr1 is a key regulator of the trunk to tail transition. Tgfbr1 has been shown to control the switch of the neuromesodermal competent cells from the epiblast to the chordoneural hinge to generate the tail bud. We now show that in mouse embryos Tgfbr1 signaling also controls the remodeling of the lateral plate mesoderm (LPM) and of the embryonic endoderm associated with the trunk to tail transition. In the absence of Tgfbr1, the two LPM layers do not converge at the end of the trunk, extending instead as separate layers until the caudal embryonic extremity, and failing to activate markers of primordia for the hindlimb and external genitalia. The vascular remodeling involving the dorsal aorta and the umbilical artery leading to the connection between embryonic and extraembryonic circulation was also affected in the Tgfbr1 mutant embryos. Similar alterations in the LPM and vascular system were also observed in Isl1 null mutants, indicating that this factor acts in the regulatory cascade downstream of Tgfbr1 in LPM-derived tissues. In addition, in the absence of Tgfbr1 the embryonic endoderm fails to expand to form the endodermal cloaca and to extend posteriorly to generate the tail gut. We present evidence suggesting that the remodeling activity of Tgfbr1 in the LPM and endoderm results from the control of the posterior PS fate after its regression during the trunk to tail transition. Our data, together with previously reported observations, place Tgfbr1 at the top of the regulatory processes controlling the trunk to tail transition.
Collapse
Affiliation(s)
| | - Ana Casaca
- Instituto Gulbenkian de Ciência, Rua da Quinta GrandeOeirasPortugal
- Gulbenkian Institute for Molecular Medicine, Avenida Prof. Egas MonizLisboaPortugal
| | - Ana Novoa
- Instituto Gulbenkian de Ciência, Rua da Quinta GrandeOeirasPortugal
- Gulbenkian Institute for Molecular Medicine, Avenida Prof. Egas MonizLisboaPortugal
| | - Ying-Yi Kuo
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Arnon D Jurberg
- Instituto Gulbenkian de Ciência, Rua da Quinta GrandeOeirasPortugal
| | - Gabriel G Martins
- Instituto Gulbenkian de Ciência, Rua da Quinta GrandeOeirasPortugal
- Gulbenkian Institute for Molecular Medicine, Avenida Prof. Egas MonizLisboaPortugal
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Moises Mallo
- Instituto Gulbenkian de Ciência, Rua da Quinta GrandeOeirasPortugal
- Gulbenkian Institute for Molecular Medicine, Avenida Prof. Egas MonizLisboaPortugal
| |
Collapse
|
39
|
Willis A, Jeong D, Liu Y, Lithopoulos MA, Yuzwa SA, Frankland PW, Kaplan DR, Miller FD. Single cell approaches define neural stem cell niches and identify microglial ligands that can enhance precursor-mediated oligodendrogenesis. Cell Rep 2025; 44:115194. [PMID: 39823226 DOI: 10.1016/j.celrep.2024.115194] [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: 03/22/2024] [Revised: 05/24/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025] Open
Abstract
Here, we used single cell RNA sequencing and single cell spatial transcriptomics to characterize the forebrain neural stem cell (NSC) niche under homeostatic and injury conditions. We defined the dorsal and lateral ventricular-subventricular zones (V-SVZs) as two distinct neighborhoods and showed that, after white matter injury, NSCs are activated to make oligodendrocytes dorsally for remyelination. This activation is coincident with an increase in transcriptionally distinct microglia in the dorsal V-SVZ niche. We modeled ligand-receptor interactions within this changing niche and identified two remyelination-associated microglial ligands, insulin growth factor 1 and oncostatin M, that promote precursor proliferation and oligodendrogenesis in culture. Infusion of either ligand into the lateral ventricles also enhanced oligodendrogenesis, even in the lateral V-SVZ, where NSCs normally make neuroblasts. These data support a model where gliogenesis versus neurogenesis is determined by the local NSC neighborhood and where injury-induced niche alterations promote NSC activation, local oligodendrogenesis, and likely contribute to myelin repair.
Collapse
Affiliation(s)
- Ashleigh Willis
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Danielle Jeong
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yunlong Liu
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Marissa A Lithopoulos
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Scott A Yuzwa
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Paul W Frankland
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David R Kaplan
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Freda D Miller
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
40
|
Eibach Y, Kreher S, Poetsch MS, Kho AL, Gaertner U, Clemen CS, Schröder R, Guo K, Milting H, Meder B, Potente M, Richter M, Schneider A, Meiners S, Gautel M, Braun T. The deubiquitinase USP5 prevents accumulation of protein aggregates in cardiomyocytes. SCIENCE ADVANCES 2025; 11:eado3852. [PMID: 39841822 PMCID: PMC11753375 DOI: 10.1126/sciadv.ado3852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
Protein homeostasis is crucial for maintaining cardiomyocyte (CM) function. Disruption of proteostasis results in accumulation of protein aggregates causing cardiac pathologies such as hypertrophy, dilated cardiomyopathy (DCM), and heart failure. Here, we identify ubiquitin-specific peptidase 5 (USP5) as a critical determinant of protein quality control (PQC) in CM. CM-specific loss of mUsp5 leads to the accumulation of polyubiquitin chains and protein aggregates, cardiac remodeling, and eventually DCM. USP5 interacts with key components of the proteostasis machinery, including PSMD14, and the absence of USP5 increases activity of the ubiquitin-proteasome system and autophagic flux in CMs. Cardiac-specific hUSP5 overexpression reduces pathological remodeling in pressure-overloaded mouse hearts and attenuates protein aggregate formation in titinopathy and desminopathy models. Since CMs from humans with end-stage DCM show lower USP5 levels and display accumulation of ubiquitinated protein aggregates, we hypothesize that therapeutically increased USP5 activity may reduce protein aggregates during DCM. Our findings demonstrate that USP5 is essential for ubiquitin turnover and proteostasis in mature CMs.
Collapse
Affiliation(s)
- Yvonne Eibach
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Frankfurt, Giessen, Germany
| | - Silke Kreher
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Frankfurt, Giessen, Germany
| | - Mareike S. Poetsch
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ay Lin Kho
- Randall Centre for Cell and Molecular Biophysics, King’s College London, BHF Centre of Excellence, London, UK
| | - Ulrich Gaertner
- University of Giessen, Institute of Anatomy and Cell Biology, Giessen, Germany
| | - Christoph S. Clemen
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Rolf Schröder
- Institute for Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Kai Guo
- Research Center Borstel/Leibniz Lung Center, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), 23845 Borstel, Germany
- Institute of Experimental Medicine, Christian-Albrechts University, Kiel, Germany
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, Bad Oeynhausen, Germany
| | - Benjamin Meder
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - Michael Potente
- Berlin Institute of Health (BIH) and Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Manfred Richter
- Department of Cardiac Surgery, Kerckhoff-Clinic, Bad Nauheim, Germany
| | - Andre Schneider
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Silke Meiners
- Research Center Borstel/Leibniz Lung Center, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), 23845 Borstel, Germany
- Institute of Experimental Medicine, Christian-Albrechts University, Kiel, Germany
| | - Mathias Gautel
- Randall Centre for Cell and Molecular Biophysics, King’s College London, BHF Centre of Excellence, London, UK
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Frankfurt, Giessen, Germany
| |
Collapse
|
41
|
Grommisch D, Lund H, Eenjes E, Julien A, Göritz C, Harris RA, Sandberg R, Hagemann-Jensen M, Genander M. Regionalized cell and gene signatures govern esophageal epithelial homeostasis. Dev Cell 2025; 60:320-336.e9. [PMID: 39426382 DOI: 10.1016/j.devcel.2024.09.025] [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: 02/16/2024] [Revised: 06/14/2024] [Accepted: 09/19/2024] [Indexed: 10/21/2024]
Abstract
Regionalized disease prevalence is a common feature of the gastrointestinal tract. Herein, we employed regionally resolved Smart-seq3 single-cell sequencing, generating a comprehensive cell atlas of the adult mouse esophagus. Characterizing the esophageal axis, we identify non-uniform distribution of epithelial basal cells, fibroblasts, and immune cells. In addition, we demonstrate a position-dependent, but cell subpopulation-independent, transcriptional signature, collectively generating a regionalized esophageal landscape. Combining in vivo models with organoid co-cultures, we demonstrate that proximal and distal basal progenitor cell states are functionally distinct. We find that proximal fibroblasts are more permissive for organoid growth compared with distal fibroblasts and that the immune cell profile is regionalized in two dimensions, where proximal-distal and epithelial-stromal gradients impact epithelial maintenance. Finally, we predict and verify how WNT, BMP, insulin growth factor (IGF), and neuregulin (NRG) signaling are differentially engaged along the esophageal axis. We establish a cellular and transcriptional framework for understanding esophageal regionalization, providing a functional basis for epithelial disease susceptibility.
Collapse
Affiliation(s)
- David Grommisch
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Harald Lund
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Evelien Eenjes
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Anais Julien
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Karolinska Institutet, Centre for Molecular Medicine, Karolinska Hospital, Stockholm, Sweden
| | - Rickard Sandberg
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Maria Genander
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
42
|
Cunha DM, Hernández-Pérez S, Mattila PK. Enhanced antibody responses in CD19-Cre mice. Sci Rep 2025; 15:1348. [PMID: 39779739 PMCID: PMC11711458 DOI: 10.1038/s41598-024-83954-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
CD19-Cre is an important and widely used Cre-lox model for B cell-specific genetic manipulation in murine systems. Mice carrying one allele of CD19-Cre are, at the same time, rendered heterozygote for CD19, a crucial coreceptor of the B cell antigen receptor (BCR). As a result, CD19-Cre mice exhibit diminished expression levels of CD19, with potential, yet insufficiently examined, consequences in B cell activation. Here, we report significantly heightened antibody responses upon both T-dependent (NP-KLH) and T-independent (NP-Ficoll) immunizations as well as elevated levels of basal IgM immunoglobulin levels in CD19-Cre mice. In vitro, we observed enhanced class-switch recombination and a moderate reduction in B cell proliferation upon LPS and IFNγ stimulation, yet no drastic differences in BCR signalling. Our findings warrant careful consideration in the use of CD19-Cre mouse model in B cell research.
Collapse
MESH Headings
- Animals
- Antigens, CD19/immunology
- Antigens, CD19/metabolism
- Mice
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Integrases/metabolism
- Antibody Formation/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/genetics
- Lymphocyte Activation/immunology
- Mice, Transgenic
- Immunoglobulin Class Switching
- Cell Proliferation
- Mice, Inbred C57BL
- Immunoglobulin M/immunology
Collapse
Affiliation(s)
- Diogo M Cunha
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
| | - Sara Hernández-Pérez
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
- Institute of Immunity and Transplantation (IIT), Division of Infection and Immunity, University College London, London, UK
| | - Pieta K Mattila
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, Finland.
- InFLAMES Research Flagship, University of Turku, Turku, Finland.
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland.
| |
Collapse
|
43
|
Zhang F, Fu Y, Jimenez-Cyrus D, Zhao T, Shen Y, Sun Y, Zhang Z, Wang Q, Kawaguchi R, Geschwind DH, He C, Ming GL, Song H. m 6A/YTHDF2-mediated mRNA decay targets TGF-β signaling to suppress the quiescence acquisition of early postnatal mouse hippocampal NSCs. Cell Stem Cell 2025; 32:144-156.e8. [PMID: 39476834 PMCID: PMC11698649 DOI: 10.1016/j.stem.2024.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/26/2024] [Accepted: 10/02/2024] [Indexed: 01/06/2025]
Abstract
Quiescence acquisition of proliferating neural stem cells (NSCs) is required to establish the adult NSC pool. The underlying molecular mechanisms are not well understood. Here, we showed that conditional deletion of the m6A reader Ythdf2, which promotes mRNA decay, in proliferating NSCs in the early postnatal mouse hippocampus elevated quiescence acquisition in a cell-autonomous fashion with decreased neurogenesis. Multimodal profiling of m6A modification, YTHDF2 binding, and mRNA decay in hippocampal NSCs identified shared targets in multiple transforming growth factor β (TGF-β)-signaling-pathway components, including TGF-β ligands, maturation factors, receptors, transcription regulators, and signaling regulators. Functionally, Ythdf2 deletion led to TGF-β-signaling activation in NSCs, suppression of which rescued elevated quiescence acquisition of proliferating hippocampal NSCs. Our study reveals the dynamic nature and critical roles of mRNA decay in establishing the quiescent adult hippocampal NSC pool and uncovers a distinct mode of epitranscriptomic control via co-regulation of multiple components of the same signaling pathway.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; School of Life Sciences, Nanjing University, Nanjing, PRC
| | - Yao Fu
- Department of Biology, School of Art and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennisse Jimenez-Cyrus
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ting Zhao
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yachen Shen
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yusha Sun
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qing Wang
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Riki Kawaguchi
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA; The Epigenetics Institute, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Neurosurgery, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
44
|
Triscott J, Lehner M, Benjak A, Reist M, Emerling BM, Ng CK, de Brot S, Rubin MA. Loss of PI5P4Kα Slows the Progression of a Pten Mutant Basal Cell Model of Prostate Cancer. Mol Cancer Res 2025; 23:33-45. [PMID: 39382632 PMCID: PMC7616865 DOI: 10.1158/1541-7786.mcr-24-0290] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 10/10/2024]
Abstract
Although early prostate cancer depends on the androgen receptor signaling pathway, which is predominant in luminal cells, there is much to be understood about the contribution of epithelial basal cells in cancer progression. Herein, we observe cell type-specific differences in the importance of the metabolic enzyme phosphatidylinositol 5-phosphate 4-kinase alpha (PI5P4Kα; gene name PIP4K2A) in the prostate epithelium. We report the development of a basal cell-specific genetically engineered mouse model targeting Pip4k2a alone or in combination with the tumor suppressor phosphatase and tensin homolog (Pten). PI5P4Kα is enriched in basal cells, and no major histopathologic changes were detectable following gene deletion. Notably, the combined loss of Pip4k2a slowed the development of Pten mutant mouse prostatic intraepithelial neoplasia. Through the inclusion of a lineage tracing reporter, we utilize single-cell RNA sequencing to evaluate changes resulting from in vivo downregulation of Pip4k2a and characterize cell populations influenced in the established Probasin-Cre- and cytokeratin 5-Cre-driven genetically engineered mouse model. Transcriptomic pathway analysis points toward the disruption of lipid metabolism as a mechanism for reduced tumor progression. This was functionally supported by shifts of carnitine lipids in LNCaP prostate cancer cells treated with siPIP4K2A. Overall, these data nominate PI5P4Kα as a target for PTEN mutant prostate cancer. Implications: PI5P4Kα is enriched in prostate basal cells, and its targeted loss slows the progression of a model of advanced prostate cancer.
Collapse
Affiliation(s)
- Joanna Triscott
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Marika Lehner
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Andrej Benjak
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Matthias Reist
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Brooke M. Emerling
- Cancer Metabolism and Microenvironment Program, Sanford Burnham Prebys, La Jolla, California
| | - Charlotte K.Y. Ng
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern and Inselspital, Bern, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Simone de Brot
- Bern Center for Precision Medicine, University of Bern and Inselspital, Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Mark A. Rubin
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern and Inselspital, Bern, Switzerland
| |
Collapse
|
45
|
Bakiri L, Tichet M, Marques C, Thomsen MK, Allen EA, Stolzlechner S, Cheng K, Matsuoka K, Squatrito M, Hanahan D, Wagner EF. A new effLuc/Kate dual reporter allele for tumor imaging in mice. Dis Model Mech 2025; 18:DMM052130. [PMID: 39745082 PMCID: PMC11789939 DOI: 10.1242/dmm.052130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/19/2024] [Indexed: 02/01/2025] Open
Abstract
Genetically engineered mouse models (GEMMs) are instrumental for modelling local and systemic features of complex diseases, such as cancer. Non-invasive, longitudinal cell detection and monitoring in tumors, metastases and/or the micro-environment is paramount to achieve a better spatiotemporal understanding of cancer progression and to evaluate therapies in preclinical studies. Bioluminescent and fluorescent reporters marking tumor cells or their microenvironment are valuable for non-invasive cell detection and monitoring in vivo. Here, we report the generation of a dual reporter allele allowing simultaneous bioluminescence and fluorescence detection of cells that have undergone Cre-Lox recombination in mice. The single copy knock-in allele in the permissive collagen I locus was evaluated in the context of several cancer GEMMs, where Cre expression was achieved genetically or by ectopic virus-mediated delivery. The new reporter allele was also combined with gene-targeted alleles widely used in bone, prostate, brain and pancreas cancer research, as well as with alleles inserted into the commonly used Rosa26 and collagen I loci. This allele is, therefore, a useful addition to the portfolio of reporters to help advance preclinical research.
Collapse
Affiliation(s)
- Latifa Bakiri
- Laboratory Genes and Disease, Department of Laboratory Medicine, Medical University of Vienna, Vienna (MUW), Spitalgasse 23, 1090 Vienna, Austria
| | - Mélanie Tichet
- Ludwig Institute for Cancer Research, Lausanne Branch; Swiss Institute for Experimental Cancer Research (ISREC), EPFL; Swiss Cancer Center Leman (SCCL); Agora Translational Cancer Research Center, Rue du Bugnon 25A, 1005 Lausanne, Switzerland
| | - Carolina Marques
- Seve Ballesteros Foundation Brain Tumor Group, Spanish National Cancer Research Centre, Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Martin K. Thomsen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus, Denmark
| | - Elizabeth A. Allen
- Ludwig Institute for Cancer Research, Lausanne Branch; Swiss Institute for Experimental Cancer Research (ISREC), EPFL; Swiss Cancer Center Leman (SCCL); Agora Translational Cancer Research Center, Rue du Bugnon 25A, 1005 Lausanne, Switzerland
| | - Stefanie Stolzlechner
- Laboratory Bone Cancer Metastasis, Cellular and Molecular Tumor Biology, Center for Cancer Research, Medical University of Vienna (MUW), Spitalgasse 23, 1090 Vienna, Austria
| | - Ke Cheng
- Ludwig Institute for Cancer Research, Lausanne Branch; Swiss Institute for Experimental Cancer Research (ISREC), EPFL; Swiss Cancer Center Leman (SCCL); Agora Translational Cancer Research Center, Rue du Bugnon 25A, 1005 Lausanne, Switzerland
| | - Kazuhiko Matsuoka
- Laboratory Bone Cancer Metastasis, Cellular and Molecular Tumor Biology, Center for Cancer Research, Medical University of Vienna (MUW), Spitalgasse 23, 1090 Vienna, Austria
| | - Massimo Squatrito
- Seve Ballesteros Foundation Brain Tumor Group, Spanish National Cancer Research Centre, Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Douglas Hanahan
- Ludwig Institute for Cancer Research, Lausanne Branch; Swiss Institute for Experimental Cancer Research (ISREC), EPFL; Swiss Cancer Center Leman (SCCL); Agora Translational Cancer Research Center, Rue du Bugnon 25A, 1005 Lausanne, Switzerland
| | - Erwin F. Wagner
- Laboratory Genes and Disease, Department of Laboratory Medicine, Medical University of Vienna, Vienna (MUW), Spitalgasse 23, 1090 Vienna, Austria
- Laboratory Genes and Disease, Department of Dermatology, Medical University of Vienna (MUW), Spitalgasse 23, 1090 Vienna, Austria
| |
Collapse
|
46
|
Cocas L, Fernandez G. Monosynaptic Tracing in Developing Circuits Using Modified Rabies Virus. Methods Mol Biol 2025; 2910:205-219. [PMID: 40220101 DOI: 10.1007/978-1-0716-4446-1_12] [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: 04/14/2025]
Abstract
An attenuated rabies virus that expresses fluorescent protein has made it possible to analyze retrograde (presynaptic) monosynaptic connections in vivo. Combining attenuated rabies virus with a Cre-loxP-based system to target cells in a subtype-specific fashion, it is possible to examine neuronal input in vivo onto any class of neuron, in development and in the mature brain. We describe here the methods to amplify deletion mutant, pseudotyped rabies virus, selectively target cells of interest using genetic and viral approaches, as well as the stereotaxic procedures required to target neuronal subtypes of interest in vivo.
Collapse
Affiliation(s)
- Laura Cocas
- Department of Biology, Santa Clara University, Santa Clara, CA, USA.
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| | - Gloria Fernandez
- Department of Biology, Santa Clara University, Santa Clara, CA, USA
| |
Collapse
|
47
|
Enbar T, Hickmott JW, Siu R, Gao D, Garcia-Flores E, Smart J, Casabuenas DL, Faiz M, Morshead CM. Regionally distinct GFAP promoter expression plays a role in off-target neuron expression following AAV5 transduction. Sci Rep 2024; 14:31583. [PMID: 39738170 PMCID: PMC11685643 DOI: 10.1038/s41598-024-79124-5] [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: 08/28/2024] [Accepted: 11/06/2024] [Indexed: 01/01/2025] Open
Abstract
Astrocyte to neuron reprogramming has been performed using viral delivery of neurogenic transcription factors in GFAP expressing cells. Recent reports of off-target expression in cortical neurons following adeno-associated virus (AAV) transduction to deliver the neurogenic factors have confounded our understanding of the efficacy of direct cellular reprogramming. To shed light on potential mechanisms that may underlie the neuronal off-target expression of GFAP promoter driven expression of neurogenic factors in neurons, two regionally distinct cortices were compared-the motor cortex (MC) and medial prefrontal cortex (mPFC)-and investigated: (1) the regional tropism and astrocyte transduction with an AAV5-GFAP vector, (2) the expression of Gfap in MC and mPFC neurons; and (3) material transfer between astrocytes and neurons. Using a Cre-based system (AAV5-hGFAP-Cre; Rosa26R-tdTomato reporter mice), regional differences were observed in tdTomato expression between the MC and mPFC. Interestingly, this correlated with the presence of a greater expression of Gfap mRNA in neurons in the mPFC. Additionally, intercellular material transfer of Cre and tdTomato was observed between astrocytes and neurons in both regions, albeit at very low frequencies. Our study highlights regionally distinct variation in neurons that warrants consideration when designing genetic constructs for gene therapies targeting astrocytes including astrocyte to neuron reprogramming.
Collapse
Affiliation(s)
- T Enbar
- Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - J W Hickmott
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - R Siu
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - D Gao
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - E Garcia-Flores
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, ON, M5S 3G9, Canada
| | - J Smart
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - D L Casabuenas
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
| | - M Faiz
- Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
| | - C M Morshead
- Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada.
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, ON, M5S 3G9, Canada.
| |
Collapse
|
48
|
Azeem M, Helal M, Klein-Hessling S, Serfling E, Goebeler M, Muhammad K, Kerstan A. NFATc1 Fosters Allergic Contact Dermatitis Responses by Enhancing the Induction of IL-17-Producing CD8 Cells. J Invest Dermatol 2024:S0022-202X(24)03036-7. [PMID: 39733935 DOI: 10.1016/j.jid.2024.11.014] [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: 11/13/2023] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/31/2024]
Abstract
A plethora of data supports a major role of CD4+ and CD8+ T lymphocytes for the initiation, progression, and maintenance of allergic contact dermatitis. However, in-depth understanding of the molecular mechanisms is still limited. NFATc1 plays an essential role in T-cell activation. We therefore investigated its impact on contact hypersensitivity, the mouse model for allergic contact dermatitis. The contact hypersensitivity response to 2,4,6-trinitrochlorobenzene was diminished in Nfatc1fl/flxCd4-cre mice (Nfatc1-/-) compared with that in wild-type mice and associated with a lower percentage of IL-17-producing CD8+ T (Tc17) cells in both inflamed skin and draining lymph nodes. In vitro Tc17 polarization assays revealed that Nfatc1-/- CD8+ T cells have a reduced capacity to polarize into Tc17 cells. Applying single-cell RNA sequencing, we realized that NFATc1 controls the T-cell differentiation fate. In the absence of NFATc1, CD8+ T cells favor the development of IFN-γ-secreting CD8+ T (Tc1) lymphocytes, whereas in its presence, they turn into Tc17 cells. Finally, the adoptive transfer of 2,4,6-trinitrochlorobenzene-sensitized wild-type CD8+ T cells restored the contact hypersensitivity response in naïve Nfatc1-/- mice. Our data demonstrate that NFATc1 contributes to the development of Tc17 cells and might present a promising target to alleviate CD8+ T-cell-mediated allergic responses.
Collapse
Affiliation(s)
- Muhammad Azeem
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany; Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Moutaz Helal
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Klein-Hessling
- Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Edgar Serfling
- Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Matthias Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Khalid Muhammad
- Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany; Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates.
| | - Andreas Kerstan
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
49
|
Dawson CA, Milevskiy MJG, Capaldo BD, Yip RKH, Song X, Vaillant F, Prokopuk L, Jackling FC, Smyth GK, Chen Y, Lindeman GJ, Visvader JE. Hormone-responsive progenitors have a unique identity and exhibit high motility during mammary morphogenesis. Cell Rep 2024; 43:115073. [PMID: 39700014 DOI: 10.1016/j.celrep.2024.115073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Hormone-receptor-positive (HR+) luminal cells largely mediate the response to estrogen and progesterone during mammary gland morphogenesis. However, there remains a lack of consensus on the precise nature of the precursor cells that maintain this essential HR+ lineage. Here we refine the identification of HR+ progenitors and demonstrate their unique regenerative capacity compared to mature HR+ cells. HR+ progenitors proliferate but do not expand, suggesting rapid differentiation. Subcellular resolution, 3D intravital microscopy was performed on terminal end buds (TEBs) during puberty to dissect the contribution of each luminal lineage. Surprisingly, HR+ TEB progenitors were highly elongated and motile compared to columnar HR- progenitors and static, conoid HR+ cells within ducts. This dynamic behavior was also observed in response to hormones. Development of an AI model for motility dynamics analysis highlighted stark behavioral changes in HR+ progenitors as they transitioned to mature cells. This work provides valuable insights into how progenitor behavior contributes to mammary morphogenesis.
Collapse
Affiliation(s)
- Caleb A Dawson
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Michael J G Milevskiy
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Bianca D Capaldo
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Raymond K H Yip
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia; Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Xiaoyu Song
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - François Vaillant
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Lexie Prokopuk
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Felicity C Jackling
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Yunshun Chen
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia; Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Geoffrey J Lindeman
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia; Parkville Familial Cancer Centre and Department of Medical Oncology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, VIC 3052, Australia
| | - Jane E Visvader
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia.
| |
Collapse
|
50
|
Díaz del Moral S, Wagner N, Wagner KD. The Wilms' Tumor Suppressor WT1 in Cardiomyocytes: Implications for Cardiac Homeostasis and Repair. Cells 2024; 13:2078. [PMID: 39768169 PMCID: PMC11674098 DOI: 10.3390/cells13242078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/11/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
The Wilms' tumor suppressor WT1 is essential for the development of the heart, among other organs such as the kidneys and gonads. The Wt1 gene encodes a zinc finger transcription factor that regulates proliferation, cellular differentiation processes, and apoptosis. WT1 is also involved in cardiac homeostasis and repair. In adulthood, WT1-expression levels are lower compared to those observed through development, and WT1 expression is restricted to a few cell types. However, its systemic deletion in adult mice is lethal, demonstrating that its presence is also key for organ maintenance. In response to injury, the epicardium re-activates the expression of WT1, but little is known about the roles it plays in cardiomyocytes, which are the main cell type affected after myocardial infarction. The fact that cardiomyocytes exhibit a low proliferation rate in the adult heart in mammals highlights the need to explore new approaches for cardiac regeneration. The aim of this review is to emphasize the functions carried out by WT1 in cardiomyocytes in cardiac homeostasis and heart regeneration.
Collapse
Affiliation(s)
| | | | - Kay-Dietrich Wagner
- Université Côte d’Azur, CNRS, INSERM, iBV, 06107 Nice, France; (S.D.d.M.); (N.W.)
| |
Collapse
|