201
|
Jerome AD, Sas AR, Wang Y, Hammond LA, Wen J, Atkinson JR, Webb A, Liu T, Segal BM. Cytokine polarized, alternatively activated bone marrow neutrophils drive axon regeneration. Nat Immunol 2024; 25:957-968. [PMID: 38811815 DOI: 10.1038/s41590-024-01836-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 04/11/2024] [Indexed: 05/31/2024]
Abstract
The adult central nervous system (CNS) possesses a limited capacity for self-repair. Severed CNS axons typically fail to regrow. There is an unmet need for treatments designed to enhance neuronal viability, facilitate axon regeneration and ultimately restore lost neurological functions to individuals affected by traumatic CNS injury, multiple sclerosis, stroke and other neurological disorders. Here we demonstrate that both mouse and human bone marrow neutrophils, when polarized with a combination of recombinant interleukin-4 (IL-4) and granulocyte colony-stimulating factor (G-CSF), upregulate alternative activation markers and produce an array of growth factors, thereby gaining the capacity to promote neurite outgrowth. Moreover, adoptive transfer of IL-4/G-CSF-polarized bone marrow neutrophils into experimental models of CNS injury triggered substantial axon regeneration within the optic nerve and spinal cord. These findings have far-reaching implications for the future development of autologous myeloid cell-based therapies that may bring us closer to effective solutions for reversing CNS damage.
Collapse
Affiliation(s)
- Andrew D Jerome
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- The Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Andrew R Sas
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- The Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Yan Wang
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- The Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Luke A Hammond
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- The Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Jing Wen
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Jeffrey R Atkinson
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Amy Webb
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Tom Liu
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- The Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Benjamin M Segal
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- The Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
202
|
Carabaña C, Sun W, Veludo Ramos C, Huyghe M, Perkins M, Maillot A, Journot R, Hartani F, Faraldo MM, Lloyd-Lewis B, Fre S. Spatially distinct epithelial and mesenchymal cell subsets along progressive lineage restriction in the branching embryonic mammary gland. EMBO J 2024; 43:2308-2336. [PMID: 38760574 PMCID: PMC11183262 DOI: 10.1038/s44318-024-00115-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 04/06/2024] [Accepted: 04/17/2024] [Indexed: 05/19/2024] Open
Abstract
How cells coordinate morphogenetic cues and fate specification during development remains a fundamental question in organogenesis. The mammary gland arises from multipotent stem cells (MaSCs), which are progressively replaced by unipotent progenitors by birth. However, the lack of specific markers for early fate specification has prevented the delineation of the features and spatial localization of MaSC-derived lineage-committed progenitors. Here, using single-cell RNA sequencing from E13.5 to birth, we produced an atlas of matched mouse mammary epithelium and mesenchyme and reconstructed the differentiation trajectories of MaSCs toward basal and luminal fate. We show that murine MaSCs exhibit lineage commitment just prior to the first sprouting events of mammary branching morphogenesis at E15.5. We identify early molecular markers for committed and multipotent MaSCs and define their spatial distribution within the developing tissue. Furthermore, we show that the mammary embryonic mesenchyme is composed of two spatially restricted cell populations, and that dermal mesenchyme-produced FGF10 is essential for embryonic mammary branching morphogenesis. Altogether, our data elucidate the spatiotemporal signals underlying lineage specification of multipotent MaSCs, and uncover the signals from mesenchymal cells that guide mammary branching morphogenesis.
Collapse
Affiliation(s)
- Claudia Carabaña
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, 75248, Paris, France
- Sorbonne Université, Collège Doctoral, Paris, France
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, C/Tajo, s/n, Villaviciosa de Odón, 28670, Madrid, Spain
| | - Wenjie Sun
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, 75248, Paris, France
| | - Camila Veludo Ramos
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, 75248, Paris, France
| | - Mathilde Huyghe
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, 75248, Paris, France
| | - Meghan Perkins
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, 75248, Paris, France
| | - Aurélien Maillot
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, 75248, Paris, France
| | - Robin Journot
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, 75248, Paris, France
| | - Fatima Hartani
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, 75248, Paris, France
| | - Marisa M Faraldo
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, 75248, Paris, France
| | - Bethan Lloyd-Lewis
- School of Cellular and Molecular Medicine, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD, UK.
| | - Silvia Fre
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, 75248, Paris, France.
| |
Collapse
|
203
|
Heß L, Aliar K, Grünwald BT, Griffin R, Lozan A, Knöller M, Khokha R, Brummer T, Reinheckel T. Dipeptidyl-peptidase 9 regulates the dynamics of tumorigenesis and metastasis in breast cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167133. [PMID: 38531482 DOI: 10.1016/j.bbadis.2024.167133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/27/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
The cytosolic dipeptidyl-aminopeptidase 9 (DPP9) cleaves protein N-termini post-proline or -alanine. Our analysis of DPP9 mRNA expression from the TCGA 'breast cancer' data set revealed that low/intermediate DPP9 levels are associated with poor overall survival of breast cancer patients. To unravel the impact of DPP9 on breast cancer development and progression, the transgenic MMTV-PyMT mouse model of metastasizing breast cancer was used. In addition, tissue- and time-controlled genetic deletion of DPP9 by the Cre-loxP recombination system was done. Despite a delay of tumor onset, a higher number of lung metastases were measured in DPP9-deficient mice compared to controls. In human mammary epithelial cells with oncogenic RAS pathway activation, DPP9 deficiency delayed tumorigenic transformation and accelerated TGF-β1 induced epithelial-to-mesenchymal transition (EMT) of spheroids. For further analysis of the mechanism, primary breast tumor cells were isolated from the MMTV-PyMT model. DPP9 deficiency in these cells caused cancer cell migration and invasion accompanied by EMT. In absence of DPP9, the EMT transcription factor ZEB1 was stabilized due to insufficient degradation by the proteasome. In summary, low expression of DPP9 appears to decelerate mammary tumorigenesis but favors EMT and metastasis, which establishes DPP9 as a novel dynamic regulator of breast cancer initiation and progression.
Collapse
Affiliation(s)
- Lisa Heß
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Kazeera Aliar
- Princess Margaret Cancer Centre, University Health Network, ON M5G 2G4, Toronto, Canada
| | - Barbara T Grünwald
- Princess Margaret Cancer Centre, University Health Network, ON M5G 2G4, Toronto, Canada
| | - Ricarda Griffin
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Alina Lozan
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; German Cancer Consortium (DKTK), partner site Freiburg, 79104 Freiburg, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Mariel Knöller
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Rama Khokha
- Princess Margaret Cancer Centre, University Health Network, ON M5G 2G4, Toronto, Canada; Department of Medical Biophysics, University of Toronto, ON M5G 2G4, Toronto, Canada
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; German Cancer Consortium (DKTK), partner site Freiburg, 79104 Freiburg, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Centre for Biological Signalling Studies BIOSS, University of Freiburg, 79104 Freiburg, Germany; Comprehensive Cancer Center Freiburg (CCCF), University Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; German Cancer Consortium (DKTK), partner site Freiburg, 79104 Freiburg, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Centre for Biological Signalling Studies BIOSS, University of Freiburg, 79104 Freiburg, Germany; Comprehensive Cancer Center Freiburg (CCCF), University Medical Center, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
204
|
Subramani PG, Fraszczak J, Helness A, Estall JL, Möröy T, Di Noia JM. Conserved role of hnRNPL in alternative splicing of epigenetic modifiers enables B cell activation. EMBO Rep 2024; 25:2662-2697. [PMID: 38744970 PMCID: PMC11169469 DOI: 10.1038/s44319-024-00152-3] [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: 10/05/2023] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024] Open
Abstract
The multifunctional RNA-binding protein hnRNPL is implicated in antibody class switching but its broader function in B cells is unknown. Here, we show that hnRNPL is essential for B cell activation, germinal center formation, and antibody responses. Upon activation, hnRNPL-deficient B cells show proliferation defects and increased apoptosis. Comparative analysis of RNA-seq data from activated B cells and another eight hnRNPL-depleted cell types reveals common effects on MYC and E2F transcriptional programs required for proliferation. Notably, while individual gene expression changes are cell type specific, several alternative splicing events affecting histone modifiers like KDM6A and SIRT1, are conserved across cell types. Moreover, hnRNPL-deficient B cells show global changes in H3K27me3 and H3K9ac. Epigenetic dysregulation after hnRNPL loss could underlie differential gene expression and upregulation of lncRNAs, and explain common and cell type-specific phenotypes, such as dysfunctional mitochondria and ROS overproduction in mouse B cells. Thus, hnRNPL is essential for the resting-to-activated B cell transition by regulating transcriptional programs and metabolism, at least in part through the alternative splicing of several histone modifiers.
Collapse
Affiliation(s)
- Poorani Ganesh Subramani
- Institut de Recherches Cliniques de Montréal, 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Boulevard Decarie, Montreal, QC, H4A 3J1, Canada
| | - Jennifer Fraszczak
- Institut de Recherches Cliniques de Montréal, 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
| | - Anne Helness
- Institut de Recherches Cliniques de Montréal, 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
| | - Jennifer L Estall
- Institut de Recherches Cliniques de Montréal, 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Boulevard Decarie, Montreal, QC, H4A 3J1, Canada
- Molecular Biology Programs, Université de Montréal, C.P. 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
- Department of Medicine, Université de Montréal, C.P. 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - Tarik Möröy
- Institut de Recherches Cliniques de Montréal, 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Boulevard Decarie, Montreal, QC, H4A 3J1, Canada
- Molecular Biology Programs, Université de Montréal, C.P. 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, 2900 Boul Edouard-Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Javier M Di Noia
- Institut de Recherches Cliniques de Montréal, 110 avenue des Pins Ouest, Montréal, QC, H2W 1R7, Canada.
- Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Boulevard Decarie, Montreal, QC, H4A 3J1, Canada.
- Molecular Biology Programs, Université de Montréal, C.P. 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada.
- Department of Medicine, Université de Montréal, C.P. 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada.
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, 2900 Boul Edouard-Montpetit, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
205
|
Kuriki M, Korb A, Comai G, Tajbakhsh S. Interplay between Pitx2 and Pax7 temporally governs specification of extraocular muscle stem cells. PLoS Genet 2024; 20:e1010935. [PMID: 38875306 PMCID: PMC11178213 DOI: 10.1371/journal.pgen.1010935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/05/2024] [Indexed: 06/16/2024] Open
Abstract
Gene regulatory networks that act upstream of skeletal muscle fate determinants are distinct in different anatomical locations. Despite recent efforts, a clear understanding of the cascade of events underlying the emergence and maintenance of the stem cell pool in specific muscle groups remains unresolved and debated. Here, we invalidated Pitx2 with multiple Cre-driver mice prenatally, postnatally, and during lineage progression. We showed that this gene becomes progressively dispensable for specification and maintenance of the muscle stem (MuSC) cell pool in extraocular muscles (EOMs) despite being, together with Myf5, a major upstream regulator during early development. Moreover, constitutive inactivation of Pax7 postnatally led to a greater loss of MuSCs in the EOMs compared to the limb. Thus, we propose a relay between Pitx2, Myf5 and Pax7 for EOM stem cell maintenance. We demonstrate also that MuSCs in the EOMs adopt a quiescent state earlier that those in limb muscles and do not spontaneously proliferate in the adult, yet EOMs have a significantly higher content of Pax7+ MuSCs per area pre- and post-natally. Finally, while limb MuSCs proliferate in the mdx mouse model for Duchenne muscular dystrophy, significantly less MuSCs were present in the EOMs of the mdx mouse model compared to controls, and they were not proliferative. Overall, our study provides a comprehensive in vivo characterisation of MuSC heterogeneity along the body axis and brings further insights into the unusual sparing of EOMs during muscular dystrophy.
Collapse
Affiliation(s)
- Mao Kuriki
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| | - Amaury Korb
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| | - Glenda Comai
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| | - Shahragim Tajbakhsh
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| |
Collapse
|
206
|
Gao Z, Santos RB, Rupert J, Van Drunen R, Yu Y, Eckel‐Mahan K, Kolonin MG. Endothelial-specific telomerase inactivation causes telomere-independent cell senescence and multi-organ dysfunction characteristic of aging. Aging Cell 2024; 23:e14138. [PMID: 38475941 PMCID: PMC11296101 DOI: 10.1111/acel.14138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
It has remained unclear how aging of endothelial cells (EC) contributes to pathophysiology of individual organs. Cell senescence results in part from inactivation of telomerase (TERT). Here, we analyzed mice with Tert knockout specifically in EC. Tert loss in EC induced transcriptional changes indicative of senescence and tissue hypoxia in EC and in other cells. We demonstrate that EC-Tert-KO mice have leaky blood vessels. The blood-brain barrier of EC-Tert-KO mice is compromised, and their cognitive function is impaired. EC-Tert-KO mice display reduced muscle endurance and decreased expression of enzymes responsible for oxidative metabolism. Our data indicate that Tert-KO EC have reduced mitochondrial content and function, which results in increased dependence on glycolysis. Consistent with this, EC-Tert-KO mice have metabolism changes indicative of increased glucose utilization. In EC-Tert-KO mice, expedited telomere attrition is observed for EC of adipose tissue (AT), while brain and skeletal muscle EC have normal telomere length but still display features of senescence. Our data indicate that the loss of Tert causes EC senescence in part through a telomere length-independent mechanism undermining mitochondrial function. We conclude that EC-Tert-KO mice is a model of expedited vascular senescence recapitulating the hallmarks aging, which can be useful for developing revitalization therapies.
Collapse
Affiliation(s)
- Zhanguo Gao
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Rafael Bravo Santos
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Joseph Rupert
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Rachel Van Drunen
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Kristin Eckel‐Mahan
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| |
Collapse
|
207
|
Matchett KP, Wilson-Kanamori JR, Portman JR, Kapourani CA, Fercoq F, May S, Zajdel E, Beltran M, Sutherland EF, Mackey JBG, Brice M, Wilson GC, Wallace SJ, Kitto L, Younger NT, Dobie R, Mole DJ, Oniscu GC, Wigmore SJ, Ramachandran P, Vallejos CA, Carragher NO, Saeidinejad MM, Quaglia A, Jalan R, Simpson KJ, Kendall TJ, Rule JA, Lee WM, Hoare M, Weston CJ, Marioni JC, Teichmann SA, Bird TG, Carlin LM, Henderson NC. Multimodal decoding of human liver regeneration. Nature 2024; 630:158-165. [PMID: 38693268 PMCID: PMC11153152 DOI: 10.1038/s41586-024-07376-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/02/2024] [Indexed: 05/03/2024]
Abstract
The liver has a unique ability to regenerate1,2; however, in the setting of acute liver failure (ALF), this regenerative capacity is often overwhelmed, leaving emergency liver transplantation as the only curative option3-5. Here, to advance understanding of human liver regeneration, we use paired single-nucleus RNA sequencing combined with spatial profiling of healthy and ALF explant human livers to generate a single-cell, pan-lineage atlas of human liver regeneration. We uncover a novel ANXA2+ migratory hepatocyte subpopulation, which emerges during human liver regeneration, and a corollary subpopulation in a mouse model of acetaminophen (APAP)-induced liver regeneration. Interrogation of necrotic wound closure and hepatocyte proliferation across multiple timepoints following APAP-induced liver injury in mice demonstrates that wound closure precedes hepatocyte proliferation. Four-dimensional intravital imaging of APAP-induced mouse liver injury identifies motile hepatocytes at the edge of the necrotic area, enabling collective migration of the hepatocyte sheet to effect wound closure. Depletion of hepatocyte ANXA2 reduces hepatocyte growth factor-induced human and mouse hepatocyte migration in vitro, and abrogates necrotic wound closure following APAP-induced mouse liver injury. Together, our work dissects unanticipated aspects of liver regeneration, demonstrating an uncoupling of wound closure and hepatocyte proliferation and uncovering a novel migratory hepatocyte subpopulation that mediates wound closure following liver injury. Therapies designed to promote rapid reconstitution of normal hepatic microarchitecture and reparation of the gut-liver barrier may advance new areas of therapeutic discovery in regenerative medicine.
Collapse
Affiliation(s)
- K P Matchett
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - J R Wilson-Kanamori
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - J R Portman
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - C A Kapourani
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- School of Informatics, University of Edinburgh, Edinburgh, UK
| | - F Fercoq
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - S May
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - E Zajdel
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - M Beltran
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - E F Sutherland
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - J B G Mackey
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - M Brice
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - G C Wilson
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - S J Wallace
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - L Kitto
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - N T Younger
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - R Dobie
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - D J Mole
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- University Department of Clinical Surgery, University of Edinburgh, Edinburgh, UK
| | - G C Oniscu
- Edinburgh Transplant Centre, Royal Infirmary of Edinburgh, Edinburgh, UK
- Division of Transplant Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - S J Wigmore
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- University Department of Clinical Surgery, University of Edinburgh, Edinburgh, UK
| | - P Ramachandran
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - C A Vallejos
- MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- The Alan Turing Institute, London, UK
| | - N O Carragher
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - M M Saeidinejad
- Institute for Liver and Digestive Health, University College London, London, UK
| | - A Quaglia
- Department of Cellular Pathology, Royal Free London NHS Foundation Trust, London, UK
- UCL Cancer Institute, University College London, London, UK
| | - R Jalan
- Institute for Liver and Digestive Health, University College London, London, UK
- European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain
| | - K J Simpson
- Department of Hepatology, University of Edinburgh and Scottish Liver Transplant Unit, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - T J Kendall
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - J A Rule
- Department of Internal Medicine, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - W M Lee
- Department of Internal Medicine, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - M Hoare
- Early Cancer Institute, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - C J Weston
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - J C Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK
- Wellcome Genome Campus, Wellcome Sanger Institute, Cambridge, UK
| | - S A Teichmann
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK
- Wellcome Genome Campus, Wellcome Sanger Institute, Cambridge, UK
- Department of Physics, Cavendish Laboratory, Cambridge, UK
| | - T G Bird
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - L M Carlin
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - N C Henderson
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
208
|
Du W, Verma A, Ye Q, Du W, Lin S, Yamanaka A, Klein OD, Hu JK. Myosin II mediates Shh signals to shape dental epithelia via control of cell adhesion and movement. PLoS Genet 2024; 20:e1011326. [PMID: 38857279 PMCID: PMC11192418 DOI: 10.1371/journal.pgen.1011326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 06/21/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024] Open
Abstract
The development of ectodermal organs begins with the formation of a stratified epithelial placode that progressively invaginates into the underlying mesenchyme as the organ takes its shape. Signaling by secreted molecules is critical for epithelial morphogenesis, but how that information leads to cell rearrangement and tissue shape changes remains an open question. Using the mouse dentition as a model, we first establish that non-muscle myosin II is essential for dental epithelial invagination and show that it functions by promoting cell-cell adhesion and persistent convergent cell movements in the suprabasal layer. Shh signaling controls these processes by inducing myosin II activation via AKT. Pharmacological induction of AKT and myosin II can also rescue defects caused by the inhibition of Shh. Together, our results support a model in which the Shh signal is transmitted through myosin II to power effective cellular rearrangement for proper dental epithelial invagination.
Collapse
Affiliation(s)
- Wei Du
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Adya Verma
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Qianlin Ye
- School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Wen Du
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Sandy Lin
- School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Atsushi Yamanaka
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Ophir D. Klein
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, California, United States of America
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Jimmy K. Hu
- School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
209
|
Yadav D, Conner JA, Wang Y, Saunders TL, Ubogu EE. A novel inducible von Willebrand Factor Cre recombinase mouse strain to study microvascular endothelial cell-specific biological processes in vivo. Vascul Pharmacol 2024; 155:107369. [PMID: 38554988 DOI: 10.1016/j.vph.2024.107369] [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: 07/18/2023] [Revised: 03/17/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
Mouse models are invaluable to understanding fundamental mechanisms in vascular biology during development, in health and different disease states. Several constitutive or inducible models that selectively knockout or knock in genes in vascular endothelial cells exist; however, functional and phenotypic differences exist between microvascular and macrovascular endothelial cells in different organs. In order to study microvascular endothelial cell-specific biological processes, we developed a Tamoxifen-inducible von Willebrand Factor (vWF) Cre recombinase mouse in the SJL background. The transgene consists of the human vWF promoter with the microvascular endothelial cell-selective 734 base pair sequence to drive Cre recombinase fused to a mutant estrogen ligand-binding domain [ERT2] that requires Tamoxifen for activity (CreERT2) followed by a polyadenylation (polyA) signal. We initially observed Tamoxifen-inducible restricted bone marrow megakaryocyte and sciatic nerve microvascular endothelial cell Cre recombinase expression in offspring of a mixed strain hemizygous C57BL/6-SJL founder mouse bred with mT/mG mice, with >90% bone marrow megakaryocyte expression efficiency. Founder mouse offspring were backcrossed to the SJL background by speed congenics, and intercrossed for >10 generations to develop hemizygous Tamoxifen-inducible vWF Cre recombinase (vWF-iCre/+) SJL mice with stable transgene insertion in chromosome 1. Microvascular endothelial cell-specific Cre recombinase expression occurred in the sciatic nerves, brains, spleens, kidneys and gastrocnemius muscles of adult vWF-iCre/+ SJL mice bred with Ai14 mice, with retained low level bone marrow and splenic megakaryocyte expression. This novel mouse strain would support hypothesis-driven mechanistic studies to decipher the role(s) of specific genes transcribed by microvascular endothelial cells during development, as well as in physiologic and pathophysiologic states in an organ- and time-dependent manner.
Collapse
Affiliation(s)
- Dinesh Yadav
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeremy A Conner
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yimin Wang
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Thomas L Saunders
- Transgenic Animal Model Core, University of Michigan, Ann Arbor, MI, USA
| | - Eroboghene E Ubogu
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
210
|
Jones Lipinski RA, Stancill JS, Nuñez R, Wynia-Smith SL, Sprague DJ, Nord JA, Bird A, Corbett JA, Smith BC. Zinc-chelating BET bromodomain inhibitors equally target islet endocrine cell types. Am J Physiol Regul Integr Comp Physiol 2024; 326:R515-R527. [PMID: 38618911 PMCID: PMC11381023 DOI: 10.1152/ajpregu.00259.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/19/2024] [Accepted: 04/07/2024] [Indexed: 04/16/2024]
Abstract
Inhibition of the bromodomain and extraterminal domain (BET) protein family is a potential strategy to prevent and treat diabetes; however, the clinical use of BET bromodomain inhibitors (BETis) is associated with adverse effects. Here, we explore a strategy for targeting BETis to β cells by exploiting the high-zinc (Zn2+) concentration in β cells relative to other cell types. We report the synthesis of a novel, Zn2+-chelating derivative of the pan-BETi (+)-JQ1, (+)-JQ1-DPA, in which (+)-JQ1 was conjugated to dipicolyl amine (DPA). As controls, we synthesized (+)-JQ1-DBA, a non-Zn2+-chelating derivative, and (-)-JQ1-DPA, an inactive enantiomer that chelates Zn2+. Molecular modeling and biophysical assays showed that (+)-JQ1-DPA and (+)-JQ1-DBA retain potent binding to BET bromodomains in vitro. Cellular assays demonstrated (+)-JQ1-DPA attenuated NF-ĸB target gene expression in β cells stimulated with the proinflammatory cytokine interleukin 1β. To assess β-cell selectivity, we isolated islets from a mouse model that expresses green fluorescent protein in insulin-positive β cells and mTomato in insulin-negative cells (non-β cells). Surprisingly, Zn2+ chelation did not confer β-cell selectivity as (+)-JQ1-DPA was equally effective in both β and α cells; however, (+)-JQ1-DPA was less effective in macrophages, a nonendocrine islet cell type. Intriguingly, the non-Zn2+-chelating derivative (+)-JQ1-DBA displayed the opposite selectivity, with greater effect in macrophages compared with (+)-JQ1-DPA, suggesting potential as a macrophage-targeting molecule. These findings suggest that Zn2+-chelating small molecules confer endocrine cell selectivity rather than β-cell selectivity in pancreatic islets and provide valuable insights and techniques to assess Zn2+ chelation as an approach to selectively target small molecules to pancreatic β cells.NEW & NOTEWORTHY Inhibition of BET bromodomains is a novel potential strategy to prevent and treat diabetes mellitus. However, BET inhibitors have negative side effects. We synthesized a BET inhibitor expected to exploit the high zinc concentration in β cells to accumulate in β cells. We show our inhibitor targeted pancreatic endocrine cells; however, it was less effective in immune cells. A control inhibitor showed the opposite effect. These findings help us understand how to target specific cells in diabetes treatment.
Collapse
Affiliation(s)
- Rachel A Jones Lipinski
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Jennifer S Stancill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Raymundo Nuñez
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Sarah L Wynia-Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Daniel J Sprague
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Joshua A Nord
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Amir Bird
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Brian C Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
211
|
McKay EJ, Luijten I, Weng X, Martinez de Morentin PB, De Frutos González E, Gao Z, Kolonin MG, Heisler LK, Semple RK. Mesenchymal-specific Alms1 knockout in mice recapitulates metabolic features of Alström syndrome. Mol Metab 2024; 84:101933. [PMID: 38583571 PMCID: PMC11047791 DOI: 10.1016/j.molmet.2024.101933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/01/2024] [Indexed: 04/09/2024] Open
Abstract
OBJECTIVE Alström Syndrome (AS), caused by biallelic ALMS1 mutations, includes obesity with disproportionately severe insulin resistant diabetes, dyslipidemia, and fatty liver. Prior studies suggest that hyperphagia is accounted for by loss of ALMS1 function in hypothalamic neurones, whereas disproportionate metabolic complications may be due to impaired adipose tissue expandability. We tested this by comparing the metabolic effects of global and mesenchymal stem cell (MSC)-specific Alms1 knockout. METHODS Global Alms1 knockout (KO) mice were generated by crossing floxed Alms1 and CAG-Cre mice. A Pdgfrα-Cre driver was used to abrogate Alms1 function selectively in MSCs and their descendants, including preadipocytes. We combined metabolic phenotyping of global and Pdgfrα+ Alms1-KO mice on a 45% fat diet with measurements of body composition and food intake, and histological analysis of metabolic tissues. RESULTS Assessed on 45% fat diet to promote adipose expansion, global Alms1 KO caused hyperphagia, obesity, insulin resistance, dyslipidaemia, and fatty liver. Pdgfrα-cre driven KO of Alms1 (MSC KO) recapitulated insulin resistance, fatty liver, and dyslipidaemia in both sexes. Other phenotypes were sexually dimorphic: increased fat mass was only present in female Alms1 MSC KO mice. Hyperphagia was not evident in male Alms1 MSC KO mice, but was found in MSC KO females, despite no neuronal Pdgfrα expression. CONCLUSIONS Mesenchymal deletion of Alms1 recapitulates metabolic features of AS, including fatty liver. This confirms a key role for Alms1 in the adipose lineage, where its loss is sufficient to cause systemic metabolic effects and damage to remote organs. Hyperphagia in females may depend on Alms1 deficiency in oligodendrocyte precursor cells rather than neurones. AS should be regarded as a forme fruste of lipodystrophy.
Collapse
Affiliation(s)
- Eleanor J McKay
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Ineke Luijten
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Xiong Weng
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Pablo B Martinez de Morentin
- The Rowett Institute, University of Aberdeen, Aberdeen, UK; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Elvira De Frutos González
- The Rowett Institute, University of Aberdeen, Aberdeen, UK; Área de Fisiología Humana, Departamento de Ciencias básicas de la Salud, Facultad de ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcón, Madrid, Spain
| | - Zhanguo Gao
- Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Mikhail G Kolonin
- Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Lora K Heisler
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Robert K Semple
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK; MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
212
|
Papinska JA, Durślewicz J, Bagavant H, Deshmukh US. Deleting Mitochondrial Superoxide Dismutase 2 in Salivary Gland Ductal Epithelial Cells Recapitulates Non-Sjögren's Sicca Syndrome. Int J Mol Sci 2024; 25:5983. [PMID: 38892170 PMCID: PMC11172772 DOI: 10.3390/ijms25115983] [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: 04/09/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Elevated oxidative stress can play a pivotal role in autoimmune diseases by exacerbating inflammatory responses and tissue damage. In Sjögren's disease (SjD), the contribution of oxidative stress in the disease pathogenesis remains unclear. To address this question, we created mice with a tamoxifen-inducible conditional knockout (KO) of a critical antioxidant enzyme, superoxide dismutase 2 (Sod2), in the salivary glands (i-sg-Sod2 KO mice). Following tamoxifen treatment, Sod2 deletion occurred primarily in the ductal epithelium, and the salivary glands showed a significant downregulation of Sod2 expression. At twelve weeks post-treatment, salivary glands from the i-sg-Sod2 KO mice exhibited increased 3-Nitrotyrosine staining. Bulk RNA-seq revealed alterations in gene expression pathways related to ribosome biogenesis, mitochondrial function, and oxidative phosphorylation. Significant changes were noted in genes characteristic of salivary gland ionocytes. The i-sg-Sod2 KO mice developed reversible glandular hypofunction. However, this functional loss was not accompanied by glandular lymphocytic foci or circulating anti-nuclear antibodies. These data demonstrate that although localized oxidative stress in salivary gland ductal cells was insufficient for SjD development, it induced glandular dysfunction. The i-sg-Sod2 KO mouse resembles patients classified as non-Sjögren's sicca and will be a valuable model for deciphering oxidative-stress-mediated glandular dysfunction and recovery mechanisms.
Collapse
Affiliation(s)
- Joanna A. Papinska
- Department of Microbiology and Immunology, Oklahoma University Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Justyna Durślewicz
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (J.D.); (H.B.)
| | - Harini Bagavant
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (J.D.); (H.B.)
| | - Umesh S. Deshmukh
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (J.D.); (H.B.)
| |
Collapse
|
213
|
Granger K, Fitch S, Shen M, Lloyd J, Bhurke A, Hancock J, Ye X, Arora R. Murine uterine gland branching is necessary for gland function in implantation. Mol Hum Reprod 2024; 30:gaae020. [PMID: 38788747 PMCID: PMC11176042 DOI: 10.1093/molehr/gaae020] [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: 10/06/2023] [Revised: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Uterine glands are branched, tubular structures whose secretions are essential for pregnancy success. It is known that pre-implantation glandular expression of leukemia inhibitory factor (LIF) is crucial for embryo implantation; however, the contribution of uterine gland structure to gland secretions, such as LIF, is not known. Here, we use mice deficient in estrogen receptor 1 (ESR1) signaling to uncover the role of ESR1 signaling in gland branching and the role of a branched structure in LIF secretion and embryo implantation. We observed that deletion of ESR1 in neonatal uterine epithelium, stroma, and muscle using the progesterone receptor PgrCre causes a block in uterine gland development at the gland bud stage. Embryonic epithelial deletion of ESR1 using a Müllerian duct Cre line, Pax2Cre, displays gland bud elongation but a failure in gland branching. Reduction of ESR1 in adult uterine epithelium using the lactoferrin-Cre (LtfCre) displays normally branched uterine glands. Unbranched glands from Pax2Cre Esr1flox/flox uteri fail to express glandular pre-implantation Lif, preventing implantation chamber formation and embryo alignment along the uterine mesometrial-antimesometrial axis. In contrast, branched glands from LtfCre Esr1flox/flox uteri display reduced expression of ESR1 and glandular Lif resulting in delayed implantation chamber formation and embryo-uterine axes alignment but mice deliver a normal number of pups. Finally, pre-pubertal unbranched glands in control mice express Lif in the luminal epithelium but fail to express Lif in the glandular epithelium, even in the presence of estrogen. These data strongly suggest that branched glands are necessary for pre-implantation glandular Lif expression for implantation success. Our study is the first to identify a relationship between the branched structure and secretory function of uterine glands and provides a framework for understanding how uterine gland structure-function contributes to pregnancy success.
Collapse
Affiliation(s)
- Katrina Granger
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Sarah Fitch
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - May Shen
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Jarrett Lloyd
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Aishwarya Bhurke
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Jonathan Hancock
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA
| | - Xiaoqin Ye
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA
| | - Ripla Arora
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
214
|
Fang Y, Yuan C, Li C, Lu C, Yu W, Wang G. The Mediator Med23 controls a transcriptional switch for muscle stem cell proliferation and differentiation in muscle regeneration. Cell Rep 2024; 43:114177. [PMID: 38691453 DOI: 10.1016/j.celrep.2024.114177] [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: 03/28/2023] [Revised: 03/14/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024] Open
Abstract
Muscle stem cells (MuSCs) contribute to a robust muscle regeneration process after injury, which is highly orchestrated by the sequential expression of multiple key transcription factors. However, it remains unclear how key transcription factors and cofactors such as the Mediator complex cooperate to regulate myogenesis. Here, we show that the Mediator Med23 is critically important for MuSC-mediated muscle regeneration. Med23 is increasingly expressed in activated/proliferating MuSCs on isolated myofibers or in response to muscle injury. Med23 deficiency reduced MuSC proliferation and enhanced its precocious differentiation, ultimately compromising muscle regeneration. Integrative analysis revealed that Med23 oppositely impacts Ternary complex factor (TCF)-targeted MuSC proliferation genes and myocardin-related transcription factor (MRTF)-targeted myogenic differentiation genes. Consistently, Med23 deficiency decreases the ETS-like transcription factor 1 (Elk1)/serum response factor (SRF) binding at proliferation gene promoters but promotes MRTF-A/SRF binding at myogenic gene promoters. Overall, our study reveals the important transcriptional control mechanism of Med23 in balancing MuSC proliferation and differentiation in muscle regeneration.
Collapse
Affiliation(s)
- Yi Fang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China; State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Chunlei Yuan
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Chonghui Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China; State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Chengjiang Lu
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China; State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Gang Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China.
| |
Collapse
|
215
|
Royo F, Garcia-Vallicrosa C, Azparren-Angulo M, Bordanaba-Florit G, Lopez-Sarrio S, Falcon-Perez JM. Three-Dimensional Hepatocyte Spheroids: Model for Assessing Chemotherapy in Hepatocellular Carcinoma. Biomedicines 2024; 12:1200. [PMID: 38927406 PMCID: PMC11201042 DOI: 10.3390/biomedicines12061200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Three-dimensional cellular models provide a more comprehensive representation of in vivo cell properties, encompassing physiological characteristics and drug susceptibility. METHODS Primary hepatocytes were seeded in ultra-low attachment plates to form spheroids, with or without tumoral cells. Spheroid structure, cell proliferation, and apoptosis were analyzed using histological staining techniques. In addition, extracellular vesicles were isolated from conditioned media by differential ultracentrifugation. Spheroids were exposed to cytotoxic drugs, and both spheroid growth and cell death were measured by microscopic imaging and flow cytometry with vital staining, respectively. RESULTS Concerning spheroid structure, an active outer layer forms a boundary with the media, while the inner core comprises a mass of cell debris. Hepatocyte-formed spheroids release vesicles into the extracellular media, and a decrease in the concentration of vesicles in the culture media can be observed over time. When co-cultured with tumoral cells, a distinct distribution pattern emerges over the primary hepatocytes, resulting in different spheroid conformations. Tumoral cell growth was compromised upon antitumoral drug challenges. CONCLUSIONS Treatment of mixed spheroids with different cytotoxic drugs enables the characterization of drug effects on both hepatocytes and tumoral cells, determining drug specificity effects on these cell types.
Collapse
Affiliation(s)
- Felix Royo
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Clara Garcia-Vallicrosa
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
| | - Maria Azparren-Angulo
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
| | - Guillermo Bordanaba-Florit
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
| | - Silvia Lopez-Sarrio
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
| | - Juan Manuel Falcon-Perez
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (M.A.-A.); (G.B.-F.); (S.L.-S.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
216
|
Wang L, Yi S, Cui X, Guo Z, Wang M, Kou X, Zhao Y, Wang H, Jiang C, Gao S, Yang G, Chen J, Gao R. Chromatin landscape instructs precise transcription factor regulome during embryonic lineage specification. Cell Rep 2024; 43:114136. [PMID: 38643480 DOI: 10.1016/j.celrep.2024.114136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/10/2024] [Accepted: 04/08/2024] [Indexed: 04/23/2024] Open
Abstract
Embryos, originating from fertilized eggs, undergo continuous cell division and differentiation, accompanied by dramatic changes in transcription, translation, and metabolism. Chromatin regulators, including transcription factors (TFs), play indispensable roles in regulating these processes. Recently, the trophoblast regulator TFAP2C was identified as crucial in initiating early cell fate decisions. However, Tfap2c transcripts persist in both the inner cell mass and trophectoderm of blastocysts, prompting inquiry into Tfap2c's function in post-lineage establishment. In this study, we delineate the dynamics of TFAP2C during the mouse peri-implantation stage and elucidate its collaboration with the key lineage regulators CDX2 and NANOG. Importantly, we propose that de novo formation of H3K9me3 in the extraembryonic ectoderm during implantation antagonizes TFAP2C binding to crucial developmental genes, thereby maintaining its lineage identity. Together, these results highlight the plasticity of the chromatin environment in designating the genomic binding of highly adaptable lineage-specific TFs and regulating embryonic cell fates.
Collapse
Affiliation(s)
- Liping Wang
- Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200072, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Shanru Yi
- Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200072, China; Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Xinyu Cui
- Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200072, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200065, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Zhenxiang Guo
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Mengting Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Xiaochen Kou
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Yanhong Zhao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Hong Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Cizhong Jiang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200065, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, Tongji University, Shanghai 200120, China.
| | - Guang Yang
- Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200072, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China.
| | - Jiayu Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China.
| | - Rui Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China.
| |
Collapse
|
217
|
Chen R, Lukianova E, Schim van der Loeff I, Spegarova JS, Willet JDP, James KD, Ryder EJ, Griffin H, IJspeert H, Gajbhiye A, Lamoliatte F, Marin-Rubio JL, Woodbine L, Lemos H, Swan DJ, Pintar V, Sayes K, Ruiz-Morales ER, Eastham S, Dixon D, Prete M, Prigmore E, Jeggo P, Boyes J, Mellor A, Huang L, van der Burg M, Engelhardt KR, Stray-Pedersen A, Erichsen HC, Gennery AR, Trost M, Adams DJ, Anderson G, Lorenc A, Trynka G, Hambleton S. NUDCD3 deficiency disrupts V(D)J recombination to cause SCID and Omenn syndrome. Sci Immunol 2024; 9:eade5705. [PMID: 38787962 DOI: 10.1126/sciimmunol.ade5705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 04/24/2024] [Indexed: 05/26/2024]
Abstract
Inborn errors of T cell development present a pediatric emergency in which timely curative therapy is informed by molecular diagnosis. In 11 affected patients across four consanguineous kindreds, we detected homozygosity for a single deleterious missense variant in the gene NudC domain-containing 3 (NUDCD3). Two infants had severe combined immunodeficiency with the complete absence of T and B cells (T -B- SCID), whereas nine showed classical features of Omenn syndrome (OS). Restricted antigen receptor gene usage by residual T lymphocytes suggested impaired V(D)J recombination. Patient cells showed reduced expression of NUDCD3 protein and diminished ability to support RAG-mediated recombination in vitro, which was associated with pathologic sequestration of RAG1 in the nucleoli. Although impaired V(D)J recombination in a mouse model bearing the homologous variant led to milder immunologic abnormalities, NUDCD3 is absolutely required for healthy T and B cell development in humans.
Collapse
Affiliation(s)
- Rui Chen
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Elena Lukianova
- Wellcome Sanger Institute, Wellcome Genome Campus, CB10 1SA Hinxton, UK
| | - Ina Schim van der Loeff
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
- Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, NE1 4LP Newcastle upon Tyne, UK
| | | | - Joseph D P Willet
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Kieran D James
- Institute of Immunology and Immunotherapy, University of Birmingham. B15 2TT Birmingham, UK
| | - Edward J Ryder
- Wellcome Sanger Institute, Wellcome Genome Campus, CB10 1SA Hinxton, UK
| | - Helen Griffin
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Hanna IJspeert
- Department of Immunology, Erasmus University Medical Center, Rotterdam 3000 CA, Netherlands
| | - Akshada Gajbhiye
- Biosciences Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Frederic Lamoliatte
- Biosciences Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Jose L Marin-Rubio
- Biosciences Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Lisa Woodbine
- Genome Damage and Stability Centre, University of Sussex, BN1 9RQ Brighton, UK
| | - Henrique Lemos
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - David J Swan
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Valeria Pintar
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Kamal Sayes
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | | | - Simon Eastham
- Wellcome Sanger Institute, Wellcome Genome Campus, CB10 1SA Hinxton, UK
| | - David Dixon
- Biosciences Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, CB10 1SA Hinxton, UK
| | - Elena Prigmore
- Wellcome Sanger Institute, Wellcome Genome Campus, CB10 1SA Hinxton, UK
| | - Penny Jeggo
- Genome Damage and Stability Centre, University of Sussex, BN1 9RQ Brighton, UK
| | - Joan Boyes
- Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, UK
| | - Andrew Mellor
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Lei Huang
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Mirjam van der Burg
- Department of Immunology, Erasmus University Medical Center, Rotterdam 3000 CA, Netherlands
| | - Karin R Engelhardt
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - Asbjørg Stray-Pedersen
- Norwegian National Unit for Newborn Screening, Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo 0424, Norway
| | - Hans Christian Erichsen
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo 0424, Norway
| | - Andrew R Gennery
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
- Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, NE1 4LP Newcastle upon Tyne, UK
| | - Matthias Trost
- Biosciences Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
| | - David J Adams
- Wellcome Sanger Institute, Wellcome Genome Campus, CB10 1SA Hinxton, UK
| | - Graham Anderson
- Institute of Immunology and Immunotherapy, University of Birmingham. B15 2TT Birmingham, UK
| | - Anna Lorenc
- Wellcome Sanger Institute, Wellcome Genome Campus, CB10 1SA Hinxton, UK
| | - Gosia Trynka
- Wellcome Sanger Institute, Wellcome Genome Campus, CB10 1SA Hinxton, UK
- Open Targets, Wellcome Genome Campus, CB10 1SA Hinxton, UK
| | - Sophie Hambleton
- Translational and Clinical Research Institute, Newcastle University, NE2 4HH Newcastle upon Tyne, UK
- Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, NE1 4LP Newcastle upon Tyne, UK
| |
Collapse
|
218
|
Weinberger T, Denise M, Joppich M, Fischer M, Garcia Rodriguez C, Kumaraswami K, Wimmler V, Ablinger S, Räuber S, Fang J, Liu L, Liu WH, Winterhalter J, Lichti J, Thomas L, Esfandyari D, Percin G, Matin S, Hidalgo A, Waskow C, Engelhardt S, Todica A, Zimmer R, Pridans C, Gomez Perdiguero E, Schulz C. Resident and recruited macrophages differentially contribute to cardiac healing after myocardial ischemia. eLife 2024; 12:RP89377. [PMID: 38775664 PMCID: PMC11111219 DOI: 10.7554/elife.89377] [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: 05/24/2024] Open
Abstract
Cardiac macrophages are heterogenous in phenotype and functions, which has been associated with differences in their ontogeny. Despite extensive research, our understanding of the precise role of different subsets of macrophages in ischemia/reperfusion (I/R) injury remains incomplete. We here investigated macrophage lineages and ablated tissue macrophages in homeostasis and after I/R injury in a CSF1R-dependent manner. Genomic deletion of a fms-intronic regulatory element (FIRE) in the Csf1r locus resulted in specific absence of resident homeostatic and antigen-presenting macrophages, without affecting the recruitment of monocyte-derived macrophages to the infarcted heart. Specific absence of homeostatic, monocyte-independent macrophages altered the immune cell crosstalk in response to injury and induced proinflammatory neutrophil polarization, resulting in impaired cardiac remodeling without influencing infarct size. In contrast, continuous CSF1R inhibition led to depletion of both resident and recruited macrophage populations. This augmented adverse remodeling after I/R and led to an increased infarct size and deterioration of cardiac function. In summary, resident macrophages orchestrate inflammatory responses improving cardiac remodeling, while recruited macrophages determine infarct size after I/R injury. These findings attribute distinct beneficial effects to different macrophage populations in the context of myocardial infarction.
Collapse
Affiliation(s)
- Tobias Weinberger
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement et Cellules SouchesParisFrance
| | - Messerer Denise
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Markus Joppich
- LFE Bioinformatik, Department of Informatics, Ludwig Maximilian UniversityMunichGermany
| | - Maximilian Fischer
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
| | - Clarisabel Garcia Rodriguez
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement et Cellules SouchesParisFrance
| | - Konda Kumaraswami
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Vanessa Wimmler
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Sonja Ablinger
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Saskia Räuber
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
- Department of Neurology, Medical Faculty, Heinrich Heine University of DüsseldorfDüsseldorfGermany
| | - Jiahui Fang
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Lulu Liu
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Wing Han Liu
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
| | - Julia Winterhalter
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Johannes Lichti
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
| | - Lukas Thomas
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
| | - Dena Esfandyari
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
- Institute of Pharmacology and Toxicology, Technical University MunichMunichGermany
| | - Guelce Percin
- Immunology of Aging, Leibniz-Institute on Aging - Fritz-Lipmann-Institute (FLI)JenaGermany
| | - Sandra Matin
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos IIIMadridSpain
| | - Andrés Hidalgo
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos IIIMadridSpain
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of MedicineNew HavenUnited States
| | - Claudia Waskow
- Immunology of Aging, Leibniz-Institute on Aging - Fritz-Lipmann-Institute (FLI)JenaGermany
- Faculty of Biological Sciences, Friedrich-Schiller-UniversityJenaGermany
| | - Stefan Engelhardt
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
- Institute of Pharmacology and Toxicology, Technical University MunichMunichGermany
| | - Andrei Todica
- Department of Nuclear Medicine, Ludwig Maximilian UniversityMunichGermany
| | - Ralf Zimmer
- LFE Bioinformatik, Department of Informatics, Ludwig Maximilian UniversityMunichGermany
| | - Clare Pridans
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
- University of Edinburgh Centre for Inflammation Research, The Queen's Medical Research InstituteEdinburghUnited Kingdom
| | - Elisa Gomez Perdiguero
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement et Cellules SouchesParisFrance
| | - Christian Schulz
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
- Department of Immunopharmacology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg UniversityMannheimGermany
| |
Collapse
|
219
|
Xu J, Deng Y, Li G. Keratin 19 (Krt19) is a novel marker gene for epicardial cells. Front Genet 2024; 15:1385867. [PMID: 38831775 PMCID: PMC11145414 DOI: 10.3389/fgene.2024.1385867] [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: 02/13/2024] [Accepted: 04/26/2024] [Indexed: 06/05/2024] Open
Abstract
Epicardial cells regulate heart growth by secreting numerous growth factors and undergoing lineage specification into other cardiac lineages. However, the lack of specific marker genes for epicardial cells has hindered the understanding of this cell type in heart development. Through the analysis of a cardiac single cell mRNA sequencing dataset, we identified a novel epicardial gene named Keratin 19 (Krt19). Further analysis of the expression patterns of Krt19 and Wt1, a well-known epicardial gene, revealed their preferences in major cardiac cell types. Using lineage-tracing analysis, we analyzed Krt19-CreER labeled cells at multiple time windows and found that it labels epicardial cells at both embryonic and neonatal stages. Furthermore, we studied the function of epicardial cells using a diphtheria toxin A chain (DTA)-based cell ablation system. We discovered that Krt19-CreER labeled cells are essential for fetal heart development. Finally, we investigated the function of Krt19-CreER and Wt1-CreER labeled cells in neonatal mouse development. We observed that the Krt19-CreER; Rosa-DTA mice displayed a smaller size after tamoxifen treatment, suggesting the potential importance of Krt19-CreER labeled cells in neonatal mouse development. Additionally, we found that Wt1-CreER; Rosa-DTA mice died at early stages, likely due to defects in the kidney and spleen. In summary, we have identified Krt19 as a new epicardial cell marker gene and further explored the function of epicardial cells using the Krt19-CreER and Wt1-CreER-mediated DTA ablation system.
Collapse
Affiliation(s)
| | | | - Guang Li
- Department of Cell Biology, Center for Integrative Organ Systems, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
220
|
Krotenberg Garcia A, Ledesma-Terrón M, Lamprou M, Vriend J, van Luyk ME, Suijkerbuijk SJE. Cell competition promotes metastatic intestinal cancer through a multistage process. iScience 2024; 27:109718. [PMID: 38706869 PMCID: PMC11068562 DOI: 10.1016/j.isci.2024.109718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/28/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
Cell competition plays an instrumental role in quality control during tissue development and homeostasis. Nevertheless, cancer cells can exploit this process for their own proliferative advantage. In our study, we generated mixed murine organoids and microtissues to explore the impact of cell competition on liver metastasis. Unlike competition at the primary site, the initial effect on liver progenitor cells does not involve the induction of apoptosis. Instead, metastatic competition manifests as a multistage process. Initially, liver progenitors undergo compaction, which is followed by cell-cycle arrest, ultimately forcing differentiation. Subsequently, the newly differentiated liver cells exhibit reduced cellular fitness, rendering them more susceptible to outcompetition by intestinal cancer cells. Notably, cancer cells leverage different interactions with different epithelial populations in the liver, using them as scaffolds to facilitate their growth. Consequently, tissue-specific mechanisms of cell competition are fundamental in driving metastatic intestinal cancer.
Collapse
Affiliation(s)
- Ana Krotenberg Garcia
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Mario Ledesma-Terrón
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
- Universidad Autónoma de Madrid (UAM), University City of Cantoblanco, 28049 Madrid, Spain
| | - Maria Lamprou
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Joyce Vriend
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Merel Elise van Luyk
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Saskia Jacoba Elisabeth Suijkerbuijk
- Division of Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| |
Collapse
|
221
|
Wang T, Shao J, Kumar S, Alnouri MW, Carvalho J, Günther S, Krasel C, Murphy KT, Bünemann M, Offermanns S, Wettschureck N. Orphan GPCR GPRC5C Facilitates Angiotensin II-Induced Smooth Muscle Contraction. Circ Res 2024; 134:1259-1275. [PMID: 38597112 DOI: 10.1161/circresaha.123.323752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/29/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND GPCRs (G-protein-coupled receptors) play a central role in the regulation of smooth muscle cell (SMC) contractility, but the function of SMC-expressed orphan GPCR class C group 5 member C (GPRC5C) is unclear. The aim of this project is to define the role of GPRC5C in SMC in vitro and in vivo. METHODS We studied the role of GPRC5C in the regulation of SMC contractility and differentiation in human and murine SMC in vitro, as well as in tamoxifen-inducible, SMC-specific GPRC5C knockout mice under basal conditions and in vascular disease in vivo. RESULTS Mesenteric arteries from tamoxifen-inducible, SMC-specific GPRC5C knockout mice showed ex vivo significantly reduced angiotensin II (Ang II)-dependent calcium mobilization and contraction, whereas responses to other relaxant or contractile factors were normal. In vitro, the knockdown of GPRC5C in human aortic SMC resulted in diminished Ang II-dependent inositol phosphate production and lower myosin light chain phosphorylation. In line with this, tamoxifen-inducible, SMC-specific GPRC5C knockout mice showed reduced Ang II-induced arterial hypertension, and acute inactivation of GPRC5C was able to ameliorate established arterial hypertension. Mechanistically, we show that GPRC5C and the Ang II receptor AT1 dimerize, and knockdown of GPRC5C resulted in reduced binding of Ang II to AT1 receptors in HEK293 cells, human and murine SMC, and arteries from tamoxifen-inducible, SMC-specific GPRC5C knockout mice. CONCLUSIONS Our data show that GPRC5C regulates Ang II-dependent vascular contraction by facilitating AT1 receptor-ligand binding and signaling.
Collapse
Affiliation(s)
- Tianpeng Wang
- Department of Pharmacology (T.W., J.S., S.K., M.W.A., J.C., S.O., N.W.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jingchen Shao
- Department of Pharmacology (T.W., J.S., S.K., M.W.A., J.C., S.O., N.W.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Shamit Kumar
- Department of Pharmacology (T.W., J.S., S.K., M.W.A., J.C., S.O., N.W.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mohammad Wessam Alnouri
- Department of Pharmacology (T.W., J.S., S.K., M.W.A., J.C., S.O., N.W.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jorge Carvalho
- Department of Pharmacology (T.W., J.S., S.K., M.W.A., J.C., S.O., N.W.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform (S.G.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Cornelius Krasel
- Department of Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Germany (C.K., M.B.)
| | - Kate T Murphy
- Department of Anatomy and Physiology, The University of Melbourne, VIC, Australia (K.T.M.)
| | - Moritz Bünemann
- Department of Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Germany (C.K., M.B.)
| | - Stefan Offermanns
- Department of Pharmacology (T.W., J.S., S.K., M.W.A., J.C., S.O., N.W.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Medical Faculty, Goethe University Frankfurt, Germany (S.O., N.W.)
- German Center for Cardiovascular Research (DZHK), Frankfurt/Bad Nauheim, Germany (S.O., N.W.)
- Cardiopulmonary Institute, Frankfurt/Bad Nauheim, Germany (S.O., N.W.)
| | - Nina Wettschureck
- Department of Pharmacology (T.W., J.S., S.K., M.W.A., J.C., S.O., N.W.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Medical Faculty, Goethe University Frankfurt, Germany (S.O., N.W.)
- German Center for Cardiovascular Research (DZHK), Frankfurt/Bad Nauheim, Germany (S.O., N.W.)
- Cardiopulmonary Institute, Frankfurt/Bad Nauheim, Germany (S.O., N.W.)
| |
Collapse
|
222
|
Ramirez A, Vyzas CA, Zhao H, Eng K, Degenhardt K, Astrof S. Buffering Mechanism in Aortic Arch Artery Formation and Congenital Heart Disease. Circ Res 2024; 134:e112-e132. [PMID: 38618720 PMCID: PMC11081845 DOI: 10.1161/circresaha.123.322767] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/27/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND The resiliency of embryonic development to genetic and environmental perturbations has been long appreciated; however, little is known about the mechanisms underlying the robustness of developmental processes. Aberrations resulting in neonatal lethality are exemplified by congenital heart disease arising from defective morphogenesis of pharyngeal arch arteries (PAAs) and their derivatives. METHODS Mouse genetics, lineage tracing, confocal microscopy, and quantitative image analyses were used to investigate mechanisms of PAA formation and repair. RESULTS The second heart field (SHF) gives rise to the PAA endothelium. Here, we show that the number of SHF-derived endothelial cells (ECs) is regulated by VEGFR2 (vascular endothelial growth factor receptor 2) and Tbx1. Remarkably, when the SHF-derived EC number is decreased, PAA development can be rescued by the compensatory endothelium. Blocking such compensatory response leads to embryonic demise. To determine the source of compensating ECs and mechanisms regulating their recruitment, we investigated 3-dimensional EC connectivity, EC fate, and gene expression. Our studies demonstrate that the expression of VEGFR2 by the SHF is required for the differentiation of SHF-derived cells into PAA ECs. The deletion of 1 VEGFR2 allele (VEGFR2SHF-HET) reduces SHF contribution to the PAA endothelium, while the deletion of both alleles (VEGFR2SHF-KO) abolishes it. The decrease in SHF-derived ECs in VEGFR2SHF-HET and VEGFR2SHF-KO embryos is complemented by the recruitment of ECs from the nearby veins. Compensatory ECs contribute to PAA derivatives, giving rise to the endothelium of the aortic arch and the ductus in VEGFR2SHF-KO mutants. Blocking the compensatory response in VEGFR2SHF-KO mutants results in embryonic lethality shortly after mid-gestation. The compensatory ECs are absent in Tbx1+/- embryos, a model for 22q11 deletion syndrome, leading to unpredictable arch artery morphogenesis and congenital heart disease. Tbx1 regulates the recruitment of the compensatory endothelium in an SHF-non-cell-autonomous manner. CONCLUSIONS Our studies uncover a novel buffering mechanism underlying the resiliency of PAA development and remodeling.
Collapse
Affiliation(s)
- AnnJosette Ramirez
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Christina A. Vyzas
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Huaning Zhao
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Kevin Eng
- Department of Statistics, Rutgers University, School of Arts and Sciences, Piscataway, NJ 08854
| | - Karl Degenhardt
- Children's Hospital of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19107
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| |
Collapse
|
223
|
Zifkos K, Bochenek ML, Gogiraju R, Robert S, Pedrosa D, Kiouptsi K, Moiko K, Wagner M, Mahfoud F, Poncelet P, Münzel T, Ruf W, Reinhardt C, Panicot-Dubois L, Dubois C, Schäfer K. Endothelial PTP1B Deletion Promotes VWF Exocytosis and Venous Thromboinflammation. Circ Res 2024; 134:e93-e111. [PMID: 38563147 DOI: 10.1161/circresaha.124.324214] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Endothelial activation promotes the release of procoagulant extracellular vesicles and inflammatory mediators from specialized storage granules. Endothelial membrane exocytosis is controlled by phosphorylation. We hypothesized that the absence of PTP1B (protein tyrosine phosphatase 1B) in endothelial cells promotes venous thromboinflammation by triggering endothelial membrane fusion and exocytosis. METHODS Mice with inducible endothelial deletion of PTP1B (End.PTP1B-KO) underwent inferior vena cava ligation to induce stenosis and venous thrombosis. Primary endothelial cells from transgenic mice and human umbilical vein endothelial cells were used for mechanistic studies. RESULTS Vascular ultrasound and histology showed significantly larger venous thrombi containing higher numbers of Ly6G (lymphocyte antigen 6 family member G)-positive neutrophils in mice with endothelial PTP1B deletion, and intravital microscopy confirmed the more pronounced neutrophil recruitment following inferior vena cava ligation. RT2 PCR profiler array and immunocytochemistry analysis revealed increased endothelial activation and adhesion molecule expression in primary End.PTP1B-KO endothelial cells, including CD62P (P-selectin) and VWF (von Willebrand factor). Pretreatment with the NF-κB (nuclear factor kappa B) kinase inhibitor BAY11-7082, antibodies neutralizing CD162 (P-selectin glycoprotein ligand-1) or VWF, or arginylglycylaspartic acid integrin-blocking peptides abolished the neutrophil adhesion to End.PTP1B-KO endothelial cells in vitro. Circulating levels of annexin V+ procoagulant endothelial CD62E+ (E-selectin) and neutrophil (Ly6G+) extracellular vesicles were also elevated in End.PTP1B-KO mice after inferior vena cava ligation. Higher plasma MPO (myeloperoxidase) and Cit-H3 (citrullinated histone-3) levels and neutrophil elastase activity indicated neutrophil activation and extracellular trap formation. Infusion of End.PTP1B-KO extracellular vesicles into C57BL/6J wild-type mice most prominently enhanced the recruitment of endogenous neutrophils, and this response was blunted in VWF-deficient mice or by VWF-blocking antibodies. Reduced PTP1B binding and tyrosine dephosphorylation of SNAP23 (synaptosome-associated protein 23) resulting in increased VWF exocytosis and neutrophil adhesion were identified as mechanisms, all of which could be restored by NF-κB kinase inhibition using BAY11-7082. CONCLUSIONS Our findings show that endothelial PTP1B deletion promotes venous thromboinflammation by enhancing SNAP23 phosphorylation, endothelial VWF exocytosis, and neutrophil recruitment.
Collapse
Affiliation(s)
- Konstantinos Zifkos
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
| | - Magdalena L Bochenek
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
- Department of Cardiology, Cardiology I (M.L.B., R.G., K.M., T.M., K.S.), University Medical Center Mainz, Germany
| | - Rajinikanth Gogiraju
- Department of Cardiology, Cardiology I (M.L.B., R.G., K.M., T.M., K.S.), University Medical Center Mainz, Germany
| | - Stéphane Robert
- Aix Marseille University, National Institute of Health and Medical Research (INSERM) 1263, National Research Institute for Agriculture, Food and Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), France (S.R., L.P.-D., C.D.)
| | - Denise Pedrosa
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
| | - Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
| | - Kateryna Moiko
- Department of Cardiology, Cardiology I (M.L.B., R.G., K.M., T.M., K.S.), University Medical Center Mainz, Germany
| | - Mathias Wagner
- Institute of Pathology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany (M.W.)
| | - Felix Mahfoud
- Department of Internal Medicine III, Cardiology, Angiology and Internal Intensive Care Medicine, Saarland University Hospital and Saarland University, Homburg, Germany (F.M.)
| | | | - Thomas Münzel
- Department of Cardiology, Cardiology I (M.L.B., R.G., K.M., T.M., K.S.), University Medical Center Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (K.Z., M.L.B., D.P., K.K., W.R., C.R.), University Medical Center Mainz, Germany
| | - Laurence Panicot-Dubois
- Aix Marseille University, National Institute of Health and Medical Research (INSERM) 1263, National Research Institute for Agriculture, Food and Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), France (S.R., L.P.-D., C.D.)
| | - Christophe Dubois
- Aix Marseille University, National Institute of Health and Medical Research (INSERM) 1263, National Research Institute for Agriculture, Food and Environment (INRAE), Cardiovascular and Nutrition Research Center (C2VN), France (S.R., L.P.-D., C.D.)
| | - Katrin Schäfer
- Department of Cardiology, Cardiology I (M.L.B., R.G., K.M., T.M., K.S.), University Medical Center Mainz, Germany
| |
Collapse
|
224
|
Chen J, Ding Y, Jiang C, Qu R, Wren JD, Georgescu C, Wang X, Reuter DN, Liu B, Giles CB, Mayr CH, Schiller HB, Dai J, Stipp CS, Subramaniyan B, Wang J, Zuo H, Huang C, Fung KM, Rice HC, Sonnenberg A, Wu D, Walters MS, Zhao YY, Kanie T, Hays FA, Papin JF, Wang DW, Zhang XA. CD151 Maintains Endolysosomal Protein Quality to Inhibit Vascular Inflammation. Circ Res 2024; 134:1330-1347. [PMID: 38557119 PMCID: PMC11081830 DOI: 10.1161/circresaha.123.323190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Tetraspanin CD151 is highly expressed in endothelia and reinforces cell adhesion, but its role in vascular inflammation remains largely unknown. METHODS In vitro molecular and cellular biological analyses on genetically modified endothelial cells, in vivo vascular biological analyses on genetically engineered mouse models, and in silico systems biology and bioinformatics analyses on CD151-related events. RESULTS Endothelial ablation of Cd151 leads to pulmonary and cardiac inflammation, severe sepsis, and perilous COVID-19, and endothelial CD151 becomes downregulated in inflammation. Mechanistically, CD151 restrains endothelial release of proinflammatory molecules for less leukocyte infiltration. At the subcellular level, CD151 determines the integrity of multivesicular bodies/lysosomes and confines the production of exosomes that carry cytokines such as ANGPT2 (angiopoietin-2) and proteases such as cathepsin-D. At the molecular level, CD151 docks VCP (valosin-containing protein)/p97, which controls protein quality via mediating deubiquitination for proteolytic degradation, onto endolysosomes to facilitate VCP/p97 function. At the endolysosome membrane, CD151 links VCP/p97 to (1) IFITM3 (interferon-induced transmembrane protein 3), which regulates multivesicular body functions, to restrain IFITM3-mediated exosomal sorting, and (2) V-ATPase, which dictates endolysosome pH, to support functional assembly of V-ATPase. CONCLUSIONS Distinct from its canonical function in strengthening cell adhesion at cell surface, CD151 maintains endolysosome function by sustaining VCP/p97-mediated protein unfolding and turnover. By supporting protein quality control and protein degradation, CD151 prevents proteins from (1) buildup in endolysosomes and (2) discharge through exosomes, to limit vascular inflammation. Also, our study conceptualizes that balance between degradation and discharge of proteins in endothelial cells determines vascular information. Thus, the IFITM3/V-ATPase-tetraspanin-VCP/p97 complexes on endolysosome, as a protein quality control and inflammation-inhibitory machinery, could be beneficial for therapeutic intervention against vascular inflammation.
Collapse
Affiliation(s)
- Junxiong Chen
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Yingjun Ding
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Chao Jiang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Rongmei Qu
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | | | - Xuejun Wang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | - Beibei Liu
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Cory B. Giles
- Oklahoma Medical Research Foundation, Oklahoma City, USA
| | | | | | - Jingxing Dai
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | | | - Jie Wang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Houjuan Zuo
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Chao Huang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Kar-Ming Fung
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Heather C. Rice
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | - David Wu
- University of Chicago, Chicago, IL, USA
| | | | - You-Yang Zhao
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Tomoharu Kanie
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Franklin A. Hays
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - James F. Papin
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Dao Wen Wang
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xin A. Zhang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
- Lead contact
| |
Collapse
|
225
|
Chen Y, Zhang L, Shi X, Han J, Chen J, Zhang X, Xie D, Li Z, Niu X, Chen L, Yang C, Sun X, Zhou T, Su P, Li N, Greenblatt MB, Ke R, Huang J, Chen Z, Xu R. Characterization of the Nucleus Pulposus Progenitor Cells via Spatial Transcriptomics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303752. [PMID: 38311573 PMCID: PMC11095158 DOI: 10.1002/advs.202303752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 01/11/2024] [Indexed: 02/06/2024]
Abstract
Loss of refreshment in nucleus pulposus (NP) cellularity leads to intervertebral disc (IVD) degeneration. Nevertheless, the cellular sequence of NP cell differentiation remains unclear, although an increasing body of literature has identified markers of NP progenitor cells (NPPCs). Notably, due to their fragility, the physical enrichment of NP-derived cells has limited conventional transcriptomic approaches in multiple studies. To overcome this limitation, a spatially resolved transcriptional atlas of the mouse IVD is generated via the 10x Genomics Visium platform dividing NP spots into two clusters. Based on this, most reported NPPC-markers, including Cathepsin K (Ctsk), are rare and predominantly located within the NP-outer subset. Cell lineage tracing further evidence that a small number of Ctsk-expressing cells generate the entire adult NP tissue. In contrast, Tie2, which has long suggested labeling NPPCs, is actually neither expressed in NP subsets nor labels NPPCs and their descendants in mouse models; consistent with this, an in situ sequencing (ISS) analysis validated the absence of Tie2 in NP tissue. Similarly, no Tie2-cre-mediated labeling of NPPCs is observed in an IVD degenerative mouse model. Altogether, in this study, the first spatial transcriptomic map of the IVD is established, thereby providing a public resource for bone biology.
Collapse
Affiliation(s)
- Yu Chen
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Long Zhang
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Xueqing Shi
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Jie Han
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Jingyu Chen
- Gene Denovo Biotechnology CoGuangzhou510006China
| | - Xinya Zhang
- School of Medicine and School of Biomedical SciencesHuaqiao UniversityQuanzhou362000China
| | - Danlin Xie
- School of Medicine and School of Biomedical SciencesHuaqiao UniversityQuanzhou362000China
- School of Life SciencesWestlake UniversityHangzhou310030China
| | - Zan Li
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Xing Niu
- China Medical UniversityShenyangLiaoning110122China
| | - Lijie Chen
- China Medical UniversityShenyangLiaoning110122China
| | - Chaoyong Yang
- Department of Chemical BiologyCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Xiujie Sun
- Department of Obstetrics and GynecologySchool of MedicineXiang'an Hospital of Xiamen UniversityXiamen UniversityXiamen361102China
| | - Taifeng Zhou
- Department of Spine SurgeryGuangdong Provincial Key Laboratory of Orthopedics and TraumatologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Peiqiang Su
- Department of Spine SurgeryGuangdong Provincial Key Laboratory of Orthopedics and TraumatologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Na Li
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| | - Matthew B. Greenblatt
- Department of Pathology and Laboratory MedicineWeill Cornell Medical CollegeNew YorkNY10065USA
- Research DivisionHospital for Special SurgeryNew YorkNY10065USA
| | - Rongqin Ke
- School of Medicine and School of Biomedical SciencesHuaqiao UniversityQuanzhou362000China
| | - Jianming Huang
- Department of OrthopedicsChengong Hospital (the 73th Group Military Hospital of People's Liberation Army) affiliated to Xiamen UniversityXiamen361000China
| | - Zhe‐Sheng Chen
- College of Pharmacy and Health SciencesSt. John's UniversityNew YorkNY11439USA
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University‐ICMRS Collaborating Center for Skeletal Stem CellsState Key Laboratory of Cellular Stress BiologyFaculty of Medicine and Life SciencesSchool of MedicineXiamen UniversityXiamen361102China
- Xiamen Key Laboratory of Regeneration MedicineFujian Provincial Key Laboratory of Organ and Tissue RegenerationSchool of MedicineXiamen UniversityXiamen361102China
| |
Collapse
|
226
|
Mercier O, Quilichini PP, Magalon K, Gil F, Ghestem A, Richard F, Boudier T, Cayre M, Durbec P. Transient demyelination causes long-term cognitive impairment, myelin alteration and network synchrony defects. Glia 2024; 72:960-981. [PMID: 38363046 DOI: 10.1002/glia.24513] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024]
Abstract
In the adult brain, activity-dependent myelin plasticity is required for proper learning and memory consolidation. Myelin loss, alteration, or even subtle structural modifications can therefore compromise the network activity, leading to functional impairment. In multiple sclerosis, spontaneous myelin repair process is possible, but it is heterogeneous among patients, sometimes leading to functional recovery, often more visible at the motor level than at the cognitive level. In cuprizone-treated mouse model, massive brain demyelination is followed by spontaneous and robust remyelination. However, reformed myelin, although functional, may not exhibit the same morphological characteristics as developmental myelin, which can have an impact on the activity of neural networks. In this context, we used the cuprizone-treated mouse model to analyze the structural, functional, and cognitive long-term effects of transient demyelination. Our results show that an episode of demyelination induces despite remyelination long-term cognitive impairment, such as deficits in spatial working memory, social memory, cognitive flexibility, and hyperactivity. These deficits were associated with a reduction in myelin content in the medial prefrontal cortex (mPFC) and hippocampus (HPC), as well as structural myelin modifications, suggesting that the remyelination process may be imperfect in these structures. In vivo electrophysiological recordings showed that the demyelination episode altered the synchronization of HPC-mPFC activity, which is crucial for memory processes. Altogether, our data indicate that the myelin repair process following transient demyelination does not allow the complete recovery of the initial myelin properties in cortical structures. These subtle modifications alter network features, leading to prolonged cognitive deficits in mice.
Collapse
Affiliation(s)
- Océane Mercier
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Pascale P Quilichini
- U1106 after INS, Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Karine Magalon
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Florian Gil
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Antoine Ghestem
- U1106 after INS, Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Fabrice Richard
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Thomas Boudier
- Aix Marseille Univ, Turing Centre for Living Systems, Marseille, France
| | - Myriam Cayre
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Pascale Durbec
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| |
Collapse
|
227
|
Chen H, Xie Y, Zhang M, Huang J, Jiang W, Zhang R, Li C, Du X, Chen H, Nie Q, Liang S, Tan Q, Yang J, Jin M, Huang S, Kuang L, Su N, Qi H, Luo X, Xu X, Deng C, Chen L, Luo F. An Hsp70 promoter-based mouse for heat shock-induced gene modulation. J Mol Med (Berl) 2024; 102:693-707. [PMID: 38492027 DOI: 10.1007/s00109-024-02433-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
Physical therapy is extensively employed in clinical settings. Nevertheless, the absence of suitable animal models has resulted in an incomplete understanding of the in vivo mechanisms and cellular distribution that respond to physical stimuli. The objective of this research was to create a mouse model capable of indicating the cells affected by physical stimuli. In this study, we successfully established a mouse line based on the heat shock protein 70 (Hsp70) promoter, wherein the expression of CreERT2 can be induced by physical stimuli. Following stimulation of the mouse tail, ear, or cultured calvarias with heat shock (generated by heating, ultrasound, or laser), a distinct Cre-mediated excision was observed in cells stimulated by these physical factors with minimal occurrence of leaky reporter expression. The application of heat shock to Hsp70-CreERT2; FGFR2-P253R double transgenic mice or Hsp70-CreERT2 mice infected with AAV-BMP4 at calvarias induced the activation of Cre-dependent mutant FGFR2-P253R or BMP4 respectively, thereby facilitating the premature closure of cranial sutures or the repair of calvarial defects. This novel mouse line holds significant potential for investigating the underlying mechanisms of physical therapy, tissue repair and regeneration, lineage tracing, and targeted modulation of gene expression of cells in local tissue stimulated by physical factor at the interested time points. KEY MESSAGES: In the study, an Hsp70-CreERT2 transgenic mouse was generated for heat shock-induced gene modulation. Heat shock, ultrasound, and laser stimulation effectively activated Cre expression in Hsp70-CreERT2; reporter mice, which leads to deletion of floxed DNA sequence in the tail, ear, and cultured calvaria tissues of mice. Local laser stimuli on cultured calvarias effectively induce Fgfr2-P253R expression in Hsp70-mTmG-Fgfr2-P253R mice and result in accelerated premature closure of cranial suture. Heat shock activated AAV9-FLEX-BMP4 expression and subsequently promoted the repair of calvarial defect of Hsp70-CreERT2; Rosa26-mTmG mice.
Collapse
Affiliation(s)
- Hangang Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yangli Xie
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Mei Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Junlan Huang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Wanling Jiang
- Department of Chinese Medicine Rehabilitation, Chongqing Emergency Medical Center, Chongqing University Central Hospital), Chongqing, 400042, China
| | - Ruobin Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Can Li
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiaolan Du
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Hua Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Qiang Nie
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Sen Liang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Qiaoyan Tan
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Jing Yang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Min Jin
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Shuo Huang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Liang Kuang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Nan Su
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Huabing Qi
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Xiaoqing Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiaoling Xu
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chuxia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lin Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China.
| | - Fengtao Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
228
|
Li R, Tran DN, Lessey BA, Young SL, Kim TH, Jeong JW. Transcriptomic changes in eutopic endometrium and ectopic lesions during endometriosis progression in a mouse model. F&S SCIENCE 2024; 5:182-194. [PMID: 38342342 PMCID: PMC11116064 DOI: 10.1016/j.xfss.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 02/13/2024]
Abstract
OBJECTIVE To identify the transcriptomic changes of ectopic lesions and eutopic endometrial tissues during the progression of endometriosis, we performed transcriptomic analysis in the eutopic endometrium and ectopic lesions. DESIGN Laboratory study. SETTING Academic medical center. ANIMALS Four fertile and 4 subfertile Pgrcre/+Rosa26mTmG/+ mice with endometriosis, and 4 sham mice for each group of endometriosis mice as control. These mice underwent either surgery to induce endometriosis or sham surgery. Fertile sham and mice with endometriosis were used 1 month after surgery, whereas subfertile ones were used 3 months after surgery. INTERVENTIONS Early and chronic effects of endometriosis on transcriptomics of ectopic lesions and eutopic endometrium. MAIN OUTCOME MEASURES RNA-sequencing analysis and identification of differentially expressed genes and pathways in the ectopic lesions and eutopic uteri from mice with endometriosis and sham mice at day 3.5 of pregnancy. RESULTS Our mouse model recapitulates the transcriptomic changes of ectopic lesions in humans. RNA-sequencing analysis was performed in ectopic lesions and eutopic uteri from mice with or without endometriosis during the progression of the disease. Estrogen activity, inflammation, angiogenesis, and fibrosis pathways were consistently elevated in all the ectopic lesions compared with eutopic endometrium. Cholesterol/glucose synthesis and stem cell pluripotency pathways were more enhanced in ectopic lesions from subfertile mice compared with their eutopic endometrium. Dysregulation of infiltration of macrophage, dendritic, T and B cells was validated with the use of immunohistochemistry in ectopic lesions. Multiple ligand-receptor pairs between the ectopic and eutopic endometrium were altered compared with the sham endometrium. Suppressed WNT and EGF pathways were only found in the eutopic endometrium from subfertile not fertile mice compared with sham. CONCLUSIONS Our mouse endometriosis model recapitulates the transcriptomics of ectopic lesions in humans. Our transcriptomic analysis during endometriosis progression in our mouse model will help us understand the pathophysiology of endometriosis.
Collapse
Affiliation(s)
- Rong Li
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri
| | - Dinh Nam Tran
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri
| | - Bruce A Lessey
- Department of Obstetrics and Gynecology, Wake Forest Baptist Health, Winston-Salem, North Carolina
| | - Steven L Young
- Department of Obstetrics, Gynecology and Women's Health, Duke University, Durham, North Carolina
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri.
| |
Collapse
|
229
|
Vonada A, Wakefield L, Martinez M, Harding CO, Grompe M, Tiyaboonchai A. Complete correction of murine phenylketonuria by selection-enhanced hepatocyte transplantation. Hepatology 2024; 79:1088-1097. [PMID: 37824086 PMCID: PMC11559556 DOI: 10.1097/hep.0000000000000631] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/18/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND AND AIMS Hepatocyte transplantation for genetic liver diseases has several potential advantages over gene therapy. However, the low efficiency of cell engraftment has limited its clinical implementation. This problem could be overcome by selectively expanding transplanted donor cells until they replace enough of the liver mass to achieve therapeutic benefit. We previously described a gene therapy method to selectively expand hepatocytes deficient in cytochrome p450 reductase (Cypor) using acetaminophen (APAP). Because Cypor is required for the transformation of APAP to a hepatotoxic metabolite, Cypor-deficient cells are protected from toxicity and are able to expand following APAP-induced liver injury. Here, we apply this selection system to correct a mouse model of phenylketonuria by cell transplantation. APPROACH AND RESULTS Hepatocytes from a wild-type donor animal were edited in vitro to create Cypor deficiency and then transplanted into phenylketonuric animals. Following selection with APAP, blood phenylalanine concentrations were fully normalized and remained stable following APAP withdrawal. Cypor-deficient hepatocytes expanded from < 1% to ~14% in corrected animals, and they showed no abnormalities in blood chemistries, liver histology, or drug metabolism. CONCLUSIONS We conclude that APAP-mediated selection of transplanted hepatocytes is a potential therapeutic for phenylketonuria with long-term efficacy and a favorable safety profile.
Collapse
Affiliation(s)
- Anne Vonada
- Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Leslie Wakefield
- Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Michael Martinez
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Cary O. Harding
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Markus Grompe
- Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amita Tiyaboonchai
- Oregon Stem Cell Center, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
230
|
Horvat N, Chocarro S, Marques O, Bauer TA, Qiu R, Diaz-Jimenez A, Helm B, Chen Y, Sawall S, Sparla R, Su L, Klingmüller U, Barz M, Hentze MW, Sotillo R, Muckenthaler MU. Superparamagnetic Iron Oxide Nanoparticles Reprogram the Tumor Microenvironment and Reduce Lung Cancer Regrowth after Crizotinib Treatment. ACS NANO 2024; 18:11025-11041. [PMID: 38626916 PMCID: PMC11064219 DOI: 10.1021/acsnano.3c08335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 05/01/2024]
Abstract
ALK-positive NSCLC patients demonstrate initial responses to ALK tyrosine kinase inhibitor (TKI) treatments, but eventually develop resistance, causing rapid tumor relapse and poor survival rates. Growing evidence suggests that the combination of drug and immune therapies greatly improves patient survival; however, due to the low immunogenicity of the tumors, ALK-positive patients do not respond to currently available immunotherapies. Tumor-associated macrophages (TAMs) play a crucial role in facilitating lung cancer growth by suppressing tumoricidal immune activation and absorbing chemotherapeutics. However, they can also be programmed toward a pro-inflammatory tumor suppressive phenotype, which represents a highly active area of therapy development. Iron loading of TAMs can achieve such reprogramming correlating with an improved prognosis in lung cancer patients. We previously showed that superparamagnetic iron oxide nanoparticles containing core-cross-linked polymer micelles (SPION-CCPMs) target macrophages and stimulate pro-inflammatory activation. Here, we show that SPION-CCPMs stimulate TAMs to secrete reactive nitrogen species and cytokines that exert tumoricidal activity. We further show that SPION-CCPMs reshape the immunosuppressive Eml4-Alk lung tumor microenvironment (TME) toward a cytotoxic profile hallmarked by the recruitment of CD8+ T cells, suggesting a multifactorial benefit of SPION-CCPM application. When intratracheally instilled into lung cancer-bearing mice, SPION-CCPMs delay tumor growth and, after first line therapy with a TKI, halt the regrowth of relapsing tumors. These findings identify SPIONs-CCPMs as an adjuvant therapy, which remodels the TME, resulting in a delay in the appearance of resistant tumors.
Collapse
Affiliation(s)
- Natalie
K. Horvat
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Sara Chocarro
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Oriana Marques
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Tobias A. Bauer
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Ruiyue Qiu
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Alberto Diaz-Jimenez
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Barbara Helm
- Division
of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
| | - Yuanyuan Chen
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Stefan Sawall
- X-ray
Imaging and CT, German Cancer Research Center
(DKFZ), Im Neuenheimer
Feld 280, 69120, Heidelberg, Germany
| | - Richard Sparla
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Lu Su
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Ursula Klingmüller
- Division
of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German
Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany
| | - Matthias Barz
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
- Department
of Dermatology, University Medical Center
of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Matthias W. Hentze
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Meyerhofstr.1, 69117, Heidelberg, Germany
| | - Rocío Sotillo
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German
Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany
| | - Martina U. Muckenthaler
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site, 69120, Heidelberg/Mannheim, Germany
| |
Collapse
|
231
|
Dar A, Li A, Petrigliano FA. Lineage tracing reveals a novel PDGFRβ + satellite cell subset that contributes to myo-regeneration of chronically injured rotator cuff muscle. Sci Rep 2024; 14:9668. [PMID: 38671006 PMCID: PMC11053018 DOI: 10.1038/s41598-024-58926-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Massive rotator cuff (RC) tendon tears are associated with progressive fibro-adipogenesis and muscle atrophy that altogether cause shoulder muscle wasting. Platelet derived growth factor β (PDGFRβ) lineage cells, that co-express PDGFRα have previously been shown to directly contribute to scar formation and fat accumulation in a mouse model of irreversible tendon and nerve transection (TTDN). Conversely, PDGFRβ+ lineage cells have also been shown to be myogenic in cultures and in other models of skeletal muscle injury. We therefore hypothesized that PDGFRβ demarcates two distinct RC residing subpopulations, fibro-adipogenic and myogenic, and aimed to elucidate the identity of the PDGFRβ myogenic precursors and evaluate their contribution, if any, to RC myo-regeneration. Lineage tracing revealed increasing contribution of PDGFRβ+ myo-progenitors to the formation of GFP+ myofibers, which were the most abundant myofiber type in regenerated muscle at 2 weeks post-TTDN. Muscle regeneration preceded muscle atrophy and both advanced from the lateral site of tendon transection to the farthest medial region. GFP+/PDGFRβ+Sca-1-lin-CXCR4+Integrin-β1+ marked a novel subset of satellite cells with confirmed myogenic properties. Further studies are warranted to identify the existence of PDGFRβ+ satellite cells in human and other mouse muscles and to define their myo-regenerative potential following acute and chronic muscle injury.
Collapse
Affiliation(s)
- Ayelet Dar
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Angela Li
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Frank A Petrigliano
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
232
|
Jiang B, Ren P, He C, Wang M, Murtada SI, Chen Y, Ramachandra AB, Li G, Qin L, Assi R, Schwartz MA, Humphrey JD, Tellides G. Short-Term Disruption of TGFβ Signaling in Adult Mice Renders the Aorta Vulnerable to Hypertension-Induced Dissection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590484. [PMID: 38712205 PMCID: PMC11071440 DOI: 10.1101/2024.04.22.590484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Hypertension and transient increases in blood pressure from extreme exertion are risk factors for aortic dissection in patients with age-related vascular degeneration or inherited connective tissue disorders. Yet, the common experimental model of angiotensin II-induced aortopathy in mice appears independent of high blood pressure as lesions do not occur in response to an alternative vasoconstrictor, norepinephrine, and are not prevented by co-treatment with a vasodilator, hydralazine. We investigated vasoconstrictor administration to adult mice 1 week after disruption of TGFβ signaling in smooth muscle cells. Norepinephrine increased blood pressure and induced aortic dissection by 7 days and even within 30 minutes that was rescued by hydralazine; results were similar with angiotensin II. Changes in regulatory contractile molecule expression were not of pathological significance. Rather, reduced synthesis of extracellular matrix yielded a vulnerable aortic phenotype by decreasing medial collagen, most dynamically type XVIII, and impairing cell-matrix adhesion. We conclude that transient and sustained increases in blood pressure cause dissection in aortas rendered vulnerable by inhibition of TGFβ-driven extracellular matrix production by smooth muscle cells. A corollary is that medial fibrosis, a frequent feature of medial degeneration, may afford some protection against aortic dissection.
Collapse
|
233
|
Vishlaghi N, Guo L, Griswold-Wheeler D, Sun Y, Booker C, Crossley JL, Bancroft AC, Juan C, Korlakunta S, Ramesh S, Pagani CA, Xu L, James AW, Tower RJ, Dellinger M, Levi B. Vegfc-expressing cells form heterotopic bone after musculoskeletal injury. Cell Rep 2024; 43:114049. [PMID: 38573853 DOI: 10.1016/j.celrep.2024.114049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 02/09/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024] Open
Abstract
Heterotopic ossification (HO) is a challenging condition that occurs after musculoskeletal injury and is characterized by the formation of bone in non-skeletal tissues. While the effect of HO on blood vessels is well established, little is known about its impact on lymphatic vessels. Here, we use a mouse model of traumatic HO to investigate the relationship between HO and lymphatic vessels. We show that injury triggers lymphangiogenesis at the injury site, which is associated with elevated vascular endothelial growth factor C (VEGF-C) levels. Through single-cell transcriptomic analyses, we identify mesenchymal progenitor cells and tenocytes as sources of Vegfc. We demonstrate by lineage tracing that Vegfc-expressing cells undergo osteochondral differentiation and contribute to the formation of HO. Last, we show that Vegfc haploinsufficiency results in a nearly 50% reduction in lymphangiogenesis and HO formation. These findings shed light on the complex mechanisms underlying HO formation and its impact on lymphatic vessels.
Collapse
Affiliation(s)
- Neda Vishlaghi
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Lei Guo
- Department of Population and Data Sciences, University of Texas Southwestern, Dallas, TX, USA
| | | | - Yuxiao Sun
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Cori Booker
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Janna L Crossley
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Alec C Bancroft
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Conan Juan
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Sneha Korlakunta
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Sowmya Ramesh
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Chase A Pagani
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Lin Xu
- Department of Population and Data Sciences, University of Texas Southwestern, Dallas, TX, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Robert J Tower
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Michael Dellinger
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA.
| | - Benjamin Levi
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA.
| |
Collapse
|
234
|
Saddouk FZ, Kuzemczak A, Saito J, Greif DM. Endothelial HIFα/PDGF-B to smooth muscle Beclin1 signaling sustains pathological muscularization in pulmonary hypertension. JCI Insight 2024; 9:e162449. [PMID: 38652543 PMCID: PMC11141934 DOI: 10.1172/jci.insight.162449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
Mechanisms underlying maintenance of pathological vascular hypermuscularization are poorly delineated. Herein, we investigated retention of smooth muscle cells (SMCs) coating normally unmuscularized distal pulmonary arterioles in pulmonary hypertension (PH) mediated by chronic hypoxia with or without Sugen 5416, and reversal of this pathology. With hypoxia in mice or culture, lung endothelial cells (ECs) upregulated hypoxia-inducible factor 1α (HIF1-α) and HIF2-α, which induce platelet-derived growth factor B (PDGF-B), and these factors were reduced to normoxic levels with re-normoxia. Re-normoxia reversed hypoxia-induced pulmonary vascular remodeling, but with EC HIFα overexpression during re-normoxia, pathological changes persisted. Conversely, after establishment of distal muscularization and PH, EC-specific deletion of Hif1a, Hif2a, or Pdgfb induced reversal. In human idiopathic pulmonary artery hypertension, HIF1-α, HIF2-α, PDGF-B, and autophagy-mediating gene products, including Beclin1, were upregulated in pulmonary artery SMCs and/or lung lysates. Furthermore, in mice, hypoxia-induced EC-derived PDGF-B upregulated Beclin1 in distal arteriole SMCs, and after distal muscularization was established, re-normoxia, EC Pdgfb deletion, or treatment with STI571 (which inhibits PDGF receptors) downregulated SMC Beclin1 and other autophagy products. Finally, SMC-specific Becn1 deletion induced apoptosis, reversing distal muscularization and PH mediated by hypoxia with or without Sugen 5416. Thus, chronic hypoxia induction of the HIFα/PDGF-B axis in ECs is required for non-cell-autonomous Beclin1-mediated survival of pathological distal arteriole SMCs.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Arterioles/metabolism
- Arterioles/pathology
- Autophagy
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Beclin-1/metabolism
- Beclin-1/genetics
- Disease Models, Animal
- Endothelial Cells/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/genetics
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Indoles
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Proto-Oncogene Proteins c-sis/metabolism
- Proto-Oncogene Proteins c-sis/genetics
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pyrroles
- Signal Transduction
- Vascular Remodeling
Collapse
Affiliation(s)
- Fatima Z. Saddouk
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| | - Andrew Kuzemczak
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| | - Junichi Saito
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
235
|
Zhang Y, Kang Z, Liu M, Wang L, Liu F. Single-cell omics identifies inflammatory signaling as a trans-differentiation trigger in mouse embryos. Dev Cell 2024; 59:961-978.e7. [PMID: 38508181 DOI: 10.1016/j.devcel.2024.02.010] [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: 05/16/2023] [Revised: 01/08/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024]
Abstract
Trans-differentiation represents a direct lineage conversion; however, insufficient characterization of this process hinders its potential applications. Here, to explore a potential universal principal for trans-differentiation, we performed single-cell transcriptomic analysis of endothelial-to-hematopoietic transition (EHT), endothelial-to-mesenchymal transition, and epithelial-to-mesenchymal transition in mouse embryos. We applied three scoring indexes of entropies, cell-type signature transcription factor expression, and critical transition signals to show common features underpinning the fate plasticity of transition states. Cross-model comparison identified inflammatory-featured transition states and a common trigger role of interleukin-33 in promoting fate conversions. Multimodal profiling (integrative transcriptomic and chromatin accessibility analysis) demonstrated the inflammatory regulation of hematopoietic specification. Furthermore, multimodal omics and fate-mapping analyses showed that endothelium-specific Spi1, as an inflammatory effector, governs appropriate chromatin accessibility and transcriptional programs to safeguard EHT. Overall, our study employs single-cell omics to identify critical transition states/signals and the common trigger role of inflammatory signaling in developmental-stress-induced fate conversions.
Collapse
Affiliation(s)
- Yifan Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Zhixin Kang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute for Stem Cell and Regeneration, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Mengyao Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Feng Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China; Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute for Stem Cell and Regeneration, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
236
|
Wang Z, Wang Y, Zhou T, Chen S, Morris D, Magalhães RDM, Li M, Wang S, Wang H, Xie Y, McSwiggin H, Oliver D, Yuan S, Zheng H, Mohammed J, Lai EC, McCarrey JR, Yan W. The rapidly evolving X-linked MIR-506 family fine-tunes spermatogenesis to enhance sperm competition. eLife 2024; 13:RP90203. [PMID: 38639482 PMCID: PMC11031087 DOI: 10.7554/elife.90203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024] Open
Abstract
Despite rapid evolution across eutherian mammals, the X-linked MIR-506 family miRNAs are located in a region flanked by two highly conserved protein-coding genes (SLITRK2 and FMR1) on the X chromosome. Intriguingly, these miRNAs are predominantly expressed in the testis, suggesting a potential role in spermatogenesis and male fertility. Here, we report that the X-linked MIR-506 family miRNAs were derived from the MER91C DNA transposons. Selective inactivation of individual miRNAs or clusters caused no discernible defects, but simultaneous ablation of five clusters containing 19 members of the MIR-506 family led to reduced male fertility in mice. Despite normal sperm counts, motility, and morphology, the KO sperm were less competitive than wild-type sperm when subjected to a polyandrous mating scheme. Transcriptomic and bioinformatic analyses revealed that these X-linked MIR-506 family miRNAs, in addition to targeting a set of conserved genes, have more targets that are critical for spermatogenesis and embryonic development during evolution. Our data suggest that the MIR-506 family miRNAs function to enhance sperm competitiveness and reproductive fitness of the male by finetuning gene expression during spermatogenesis.
Collapse
Affiliation(s)
- Zhuqing Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Yue Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Sheng Chen
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Dayton Morris
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | | | - Musheng Li
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Shawn Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Hetan Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Yeming Xie
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Hayden McSwiggin
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Daniel Oliver
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Shuiqiao Yuan
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
| | - Huili Zheng
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
| | - Jaaved Mohammed
- Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Eric C Lai
- Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - John R McCarrey
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San AntonioSan AntonioUnited States
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada, Reno School of MedicineRenoUnited States
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical CenterTorranceUnited States
- Department of Medicine, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| |
Collapse
|
237
|
Chen Y, Lu X, Whitney RL, Li Y, Robson MJ, Blakely RD, Chi JT, Crowley SD, Privratsky JR. Novel anti-inflammatory effects of the IL-1 receptor in kidney myeloid cells following ischemic AKI. Front Mol Biosci 2024; 11:1366259. [PMID: 38693918 PMCID: PMC11061482 DOI: 10.3389/fmolb.2024.1366259] [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: 01/05/2024] [Accepted: 04/01/2024] [Indexed: 05/03/2024] Open
Abstract
Introduction: Acute kidney injury (AKI) is one of the most common causes of organ failure in critically ill patients. Following AKI, the canonical pro-inflammatory cytokine interleukin-1β (IL-1β) is released predominantly from activated myeloid cells and binds to the interleukin-1 receptor R1 (IL-1R1) on leukocytes and kidney parenchymal cells. IL-1R1 on kidney tubular cells is known to amplify the immune response and exacerbate AKI. However, the specific role of IL-1R1 on myeloid cells during AKI is poorly understood. The objective of the present study was to elucidate the function of myeloid cell IL-1R1 during AKI. As IL-1R1 is known to signal through the pro-inflammatory Toll-like receptor (TLR)/MyD88 pathway, we hypothesized that myeloid cells expressing IL-1R1 would exacerbate AKI. Methods: IL-1R1 was selectively depleted in CD11c+-expressing myeloid cells with CD11cCre + /IL-1R1 fl/fl (Myel KO) mice. Myel KO and littermate controls (CD11cCre - /IL-1R1 fl/fl-Myel WT) were subjected to kidney ischemia/reperfusion (I/R) injury. Kidney injury was assessed by blood urea nitrogen (BUN), serum creatinine and injury marker neutrophil gelatinase-associated lipocalin (NGAL) protein expression. Renal tubular cells (RTC) were co-cultured with CD11c+ bone marrow-derived dendritic cells (BMDC) from Myel KO and Myel WT mice. Results: Surprisingly, compared to Myel WT mice, Myel KO mice displayed exaggerated I/R-induced kidney injury, as measured by elevated levels of serum creatinine and BUN, and kidney NGAL protein expression. In support of these findings, in vitro co-culture studies showed that RTC co-cultured with Myel KO BMDC (in the presence of IL-1β) exhibited higher mRNA levels of the kidney injury marker NGAL than those co-cultured with Myel WT BMDC. In addition, we observed that IL-1R1 on Myel WT BMDC preferentially augmented the expression of anti-inflammatory cytokine interleukin-1 receptor antagonist (IL-1ra/Il1rn), effects that were largely abrogated in Myel KO BMDC. Furthermore, recombinant IL-1Ra could rescue IL-1β-induced tubular cell injury. Discussion: Our findings suggest a novel function of IL-1R1 is to serve as a critical negative feedback regulator of IL-1 signaling in CD11c+ myeloid cells to dampen inflammation to limit AKI. Our results lend further support for cell-specific, as opposed to global, targeting of immunomodulatory agents.
Collapse
Affiliation(s)
- Yanting Chen
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC, United States
| | - Xiaohan Lu
- Department of Medicine, Duke University, Durham, NC, United States
| | - Raeann L. Whitney
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC, United States
- Department of Medicine, Duke University, Durham, NC, United States
| | - Yu Li
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC, United States
- Department of Anesthesiology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi, China
| | - Matthew J. Robson
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Randy D. Blakely
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, United States
| | - Jen-Tsan Chi
- Department of Microbiology and Molecular Genetics, Duke University, Durham, NC, United States
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, United States
| | - Steven D. Crowley
- Department of Medicine, Duke University, Durham, NC, United States
- Durham VA Medical Center, Durham, NC, United States
| | - Jamie R. Privratsky
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC, United States
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, United States
| |
Collapse
|
238
|
Stewen J, Kruse K, Godoi-Filip AT, Zenia, Jeong HW, Adams S, Berkenfeld F, Stehling M, Red-Horse K, Adams RH, Pitulescu ME. Eph-ephrin signaling couples endothelial cell sorting and arterial specification. Nat Commun 2024; 15:2539. [PMID: 38570531 PMCID: PMC10991410 DOI: 10.1038/s41467-024-46300-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 02/21/2024] [Indexed: 04/05/2024] Open
Abstract
Cell segregation allows the compartmentalization of cells with similar fates during morphogenesis, which can be enhanced by cell fate plasticity in response to local molecular and biomechanical cues. Endothelial tip cells in the growing retina, which lead vessel sprouts, give rise to arterial endothelial cells and thereby mediate arterial growth. Here, we have combined cell type-specific and inducible mouse genetics, flow experiments in vitro, single-cell RNA sequencing and biochemistry to show that the balance between ephrin-B2 and its receptor EphB4 is critical for arterial specification, cell sorting and arteriovenous patterning. At the molecular level, elevated ephrin-B2 function after loss of EphB4 enhances signaling responses by the Notch pathway, VEGF and the transcription factor Dach1, which is influenced by endothelial shear stress. Our findings reveal how Eph-ephrin interactions integrate cell segregation and arteriovenous specification in the vasculature, which has potential relevance for human vascular malformations caused by EPHB4 mutations.
Collapse
Affiliation(s)
- Jonas Stewen
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Kai Kruse
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
- Bioinformatics Service Unit, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Anca T Godoi-Filip
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Zenia
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Hyun-Woo Jeong
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
- Sequencing Core Facility, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Susanne Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Frank Berkenfeld
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Martin Stehling
- Flow Cytometry Unit, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany.
| | - Mara E Pitulescu
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany.
| |
Collapse
|
239
|
Shroff NP, Xu P, Kim S, Shelton ER, Gross BJ, Liu Y, Gomez CO, Ye Q, Drennon TY, Hu JK, Green JBA, Campàs O, Klein OD. Proliferation-driven mechanical compression induces signalling centre formation during mammalian organ development. Nat Cell Biol 2024; 26:519-529. [PMID: 38570617 PMCID: PMC11482733 DOI: 10.1038/s41556-024-01380-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/15/2024] [Indexed: 04/05/2024]
Abstract
Localized sources of morphogens, called signalling centres, play a fundamental role in coordinating tissue growth and cell fate specification during organogenesis. However, how these signalling centres are established in tissues during embryonic development is still unclear. Here we show that the main signalling centre orchestrating development of rodent incisors, the enamel knot (EK), is specified by a cell proliferation-driven buildup in compressive stresses (mechanical pressure) in the tissue. Direct mechanical measurements indicate that the stresses generated by cell proliferation are resisted by the surrounding tissue, creating a circular pattern of mechanical anisotropy with a region of high compressive stress at its centre that becomes the EK. Pharmacological inhibition of proliferation reduces stresses and suppresses EK formation, and application of external pressure in proliferation-inhibited conditions rescues the formation of the EK. Mechanical information is relayed intracellularly through YAP protein localization, which is cytoplasmic in the region of compressive stress that establishes the EK and nuclear in the stretched anisotropic cells that resist the pressure buildup around the EK. Together, our data identify a new role for proliferation-driven mechanical compression in the specification of a model signalling centre during mammalian organ development.
Collapse
Affiliation(s)
- Neha Pincha Shroff
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Pengfei Xu
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Sangwoo Kim
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Institute of Mechanical Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Elijah R Shelton
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Ben J Gross
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Yucen Liu
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Carlos O Gomez
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Qianlin Ye
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Tingsheng Yu Drennon
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA, USA
| | - Jimmy K Hu
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Jeremy B A Green
- Centre for Craniofacial Regeneration and Biology, King's College London, London, UK
| | - Otger Campàs
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA.
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA.
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany.
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Center for Systems Biology Dresden, Dresden, Germany.
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA, USA.
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA.
| |
Collapse
|
240
|
Hu J, Yang F, Liu C, Wang N, Xiao Y, Zhai Y, Wang X, Zhang R, Gao L, Xu M, Wang J, Liu Z, Huang S, Liu W, Hu Y, Liu F, Guo Y, Wang L, Yuan J, Zhang Z, Chu J. UFObow: A single-wavelength excitable Brainbow for simultaneous multicolor ex-vivo and in-vivo imaging of mammalian cells. Commun Biol 2024; 7:394. [PMID: 38561421 PMCID: PMC10984974 DOI: 10.1038/s42003-024-06062-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Brainbow is a genetic cell-labeling technique that allows random colorization of multiple cells and real-time visualization of cell fate within a tissue, providing valuable insights into understanding complex biological processes. However, fluorescent proteins (FPs) in Brainbow have distinct excitation spectra with peak difference greater than 35 nm, which requires sequential imaging under multiple excitations and thus leads to long acquisition times. In addition, they are not easily used together with other fluorophores due to severe spectral bleed-through. Here, we report the development of a single-wavelength excitable Brainbow, UFObow, incorporating three newly developed blue-excitable FPs. We have demonstrated that UFObow enables not only tracking the growth dynamics of tumor cells in vivo but also mapping spatial distribution of immune cells within a sub-cubic centimeter tissue, revealing cell heterogeneity. This provides a powerful means to explore complex biology in a simultaneous imaging manner at a single-cell resolution in organs or in vivo.
Collapse
Affiliation(s)
- Jiahong Hu
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Fangfang Yang
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Chong Liu
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Nengzhi Wang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Yinghan Xiao
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yujie Zhai
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Xinru Wang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Ren Zhang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Lulu Gao
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Mengli Xu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou, Hainan, 570228, China
| | - Jialu Wang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Zheng Liu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou, Hainan, 570228, China
| | - Songlin Huang
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou, Hainan, 570228, China
| | - Wenfeng Liu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yajing Hu
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Feng Liu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yuqi Guo
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liang Wang
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jing Yuan
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China.
| | - Zhihong Zhang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China.
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou, Hainan, 570228, China.
| | - Jun Chu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Biomedical Imaging Science and System Key Laboratory, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
241
|
Greer SE, Haller SJ, Lee D, Dudley AT. N-cadherin and β1 integrin coordinately regulate growth plate cartilage architecture. Mol Biol Cell 2024; 35:ar49. [PMID: 38294852 PMCID: PMC11064670 DOI: 10.1091/mbc.e23-03-0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 12/07/2023] [Accepted: 01/23/2024] [Indexed: 02/01/2024] Open
Abstract
Spatial and temporal regulation of chondrocyte maturation in the growth plate drives growth of many bones. One essential event to generate the ordered cell array characterizing growth plate cartilage is the formation of chondrocyte columns in the proliferative zone via 90-degree rotation of daughter cells to align with the long axis of the bone. Previous studies have suggested crucial roles for cadherins and integrin β1 in column formation. The purpose of this study was to determine the relative contributions of cadherin- and integrin-mediated cell adhesion in column formation. Here we present new mechanistic insights generated by application of live time-lapse confocal microscopy of cranial base explant cultures, robust genetic mouse models, and new quantitative methods to analyze cell behavior. We show that conditional deletion of either the cell-cell adhesion molecule Cdh2 or the cell-matrix adhesion molecule Itgb1 disrupts column formation. Compound mutants were used to determine a potential reciprocal regulatory interaction between the two adhesion surfaces and identified that defective chondrocyte rotation in a N-cadherin mutant was restored by a heterozygous loss of integrin β1. Our results support a model for which integrin β1, and not N-cadherin, drives chondrocyte rotation and for which N-cadherin is a potential negative regulator of integrin β1 function.
Collapse
Affiliation(s)
- Sydney E. Greer
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198
| | - Stephen J. Haller
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198
| | - Donghee Lee
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198
| | - Andrew T. Dudley
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
242
|
Osinski V, Yellamilli A, Firulyova MM, Zhang MJ, Peck A, Auger JL, Faragher JL, Marath A, Voeller RK, O’Connell TD, Zaitsev K, Binstadt BA. Profibrotic VEGFR3-Dependent Lymphatic Vessel Growth in Autoimmune Valvular Carditis. Arterioscler Thromb Vasc Biol 2024; 44:807-821. [PMID: 38269589 PMCID: PMC10978259 DOI: 10.1161/atvbaha.123.320326] [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: 10/25/2023] [Accepted: 01/11/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Rheumatic heart disease is the major cause of valvular heart disease in developing nations. Endothelial cells (ECs) are considered crucial contributors to rheumatic heart disease, but greater insight into their roles in disease progression is needed. METHODS We used a Cdh5-driven EC lineage-tracing approach to identify and track ECs in the K/B.g7 model of autoimmune valvular carditis. Single-cell RNA sequencing was used to characterize the EC populations in control and inflamed mitral valves. Immunostaining and conventional histology were used to evaluate lineage tracing and validate single-cell RNA-sequencing findings. The effects of VEGFR3 (vascular endothelial growth factor receptor 3) and VEGF-C (vascular endothelial growth factor C) inhibitors were tested in vivo. The functional impact of mitral valve disease in the K/B.g7 mouse was evaluated using echocardiography. Finally, to translate our findings, we analyzed valves from human patients with rheumatic heart disease undergoing mitral valve replacements. RESULTS Lineage tracing in K/B.g7 mice revealed new capillary lymphatic vessels arising from valve surface ECs during the progression of disease in K/B.g7 mice. Unsupervised clustering of mitral valve single-cell RNA-sequencing data revealed novel lymphatic valve ECs that express a transcriptional profile distinct from other valve EC populations including the recently identified PROX1 (Prospero homeobox protein 1)+ lymphatic valve ECs. During disease progression, these newly identified lymphatic valve ECs expand and upregulate a profibrotic transcriptional profile. Inhibiting VEGFR3 through multiple approaches prevented expansion of this mitral valve lymphatic network. Echocardiography demonstrated that K/B.g7 mice have left ventricular dysfunction and mitral valve stenosis. Valve lymphatic density increased with age in K/B.g7 mice and correlated with worsened ventricular dysfunction. Importantly, human rheumatic valves contained similar lymphatics in greater numbers than nonrheumatic controls. CONCLUSIONS These studies reveal a novel mode of inflammation-associated, VEGFR3-dependent postnatal lymphangiogenesis in murine autoimmune valvular carditis, with similarities to human rheumatic heart disease.
Collapse
Affiliation(s)
- Victoria Osinski
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Amritha Yellamilli
- Department of Pediatrics, Stanford School of Medicine, Palo Alto, CA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN
| | - Maria M. Firulyova
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Michael J. Zhang
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Alyssa Peck
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jennifer L. Auger
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jessica L. Faragher
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| | | | | | - Timothy D. O’Connell
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
| | - Konstantin Zaitsev
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Bryce A. Binstadt
- Department of Pediatrics and Center for Immunology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
243
|
Daryadel A, Tang C, Xie Y, Peitzsch M, Fisi V, Hantel C, Loffing-Cueni D, Breault DT, Penton D, Loffing J, Beuschlein F. Zona Glomerulosa-Derived Klotho Modulates Aldosterone Synthase Expression in Young Female Mice. Endocrinology 2024; 165:bqae040. [PMID: 38573585 PMCID: PMC11002783 DOI: 10.1210/endocr/bqae040] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Klotho plays a critical role in the regulation of ion and fluid homeostasis. A previous study reported that haplo-insufficiency of Klotho in mice results in increased aldosterone synthase (CYP11B2) expression, elevated plasma aldosterone, and high blood pressure. This phenotype was presumed to be the result of diminished Klotho expression in zona glomerulosa (zG) cells of the adrenal cortex; however, systemic effects on adrenal aldosterone production could not be ruled out. To examine whether Klotho expressed in the zG is indeed a critical regulator of aldosterone synthesis, we generated a tamoxifen-inducible, zG-specific mouse model of Klotho deficiency by crossing Klotho-flox mice with Cyp11b2-CreERT mice (zG-Kl-KO). Tamoxifen-treated Cyp11b2-CreERT animals (zG-Cre) served as controls. Rosa26-mTmG reporter mice were used for Cre-dependent lineage-marking. Two weeks after tamoxifen induction, the specificity of the zG-Cre line was verified using immunofluorescence analysis to show that GFP expression was restricted to the zG. RNA in situ hybridization revealed a 65% downregulation of Klotho messenger RNA expression in the zG of zG-Kl-KO female mice at age 12 weeks compared to control mice. Despite this significant decrease, zG-Kl-KO mice exhibited no difference in plasma aldosterone levels. However, adrenal CYP11B2 expression and the CYP11B2 promotor regulatory transcription factors, NGFIB and Nurr1, were enhanced. Together with in vitro experiments, these results suggest that zG-derived Klotho modulates Cyp11b2 but does not evoke a systemic phenotype in young adult mice on a normal diet. Further studies are required to investigate the role of adrenal Klotho on aldosterone synthesis in aged animals.
Collapse
Affiliation(s)
- Arezoo Daryadel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital of Zürich (USZ) and University of Zürich (UZH), 8091 Zurich, Switzerland
| | - Cong Tang
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital of Zürich (USZ) and University of Zürich (UZH), 8091 Zurich, Switzerland
| | - Ye Xie
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital of Zürich (USZ) and University of Zürich (UZH), 8091 Zurich, Switzerland
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Viktoria Fisi
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital of Zürich (USZ) and University of Zürich (UZH), 8091 Zurich, Switzerland
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital of Zürich (USZ) and University of Zürich (UZH), 8091 Zurich, Switzerland
| | | | - David T Breault
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - David Penton
- Electrophysiology Facility, University of Zurich, 8057 Zürich, Switzerland
| | - Johannes Loffing
- Institute of Anatomy, University of Zürich, 8057 Zurich, Switzerland
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital of Zürich (USZ) and University of Zürich (UZH), 8091 Zurich, Switzerland
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität, 81377 Munich, Germany
- The LOOP Zurich Medical Research Center, 8044 Zurich, Switzerland
| |
Collapse
|
244
|
Li H, Tang Q, Yang T, Wang Z, Li D, Wang L, Li L, Chen Y, Huang H, Zhang Y, Chen Y. Segregation of morphogenetic regulatory function of Shox2 from its cell fate guardian role in sinoatrial node development. Commun Biol 2024; 7:385. [PMID: 38553636 PMCID: PMC10980793 DOI: 10.1038/s42003-024-06039-2] [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: 04/23/2023] [Accepted: 03/11/2024] [Indexed: 04/02/2024] Open
Abstract
Shox2 plays a vital role in the morphogenesis and physiological function of the sinoatrial node (SAN), the primary cardiac pacemaker, manifested by the formation of a hypoplastic SAN and failed differentiation of pacemaker cells in Shox2 mutants. Shox2 and Nkx2-5 are co-expressed in the developing SAN and regulate the fate of the pacemaker cells through a Shox2-Nkx2-5 antagonistic mechanism. Here we show that simultaneous inactivation of Nkx2-5 in the SAN of Shox2 mutants (dKO) rescued the pacemaking cell fate but not the hypoplastic defects, indicating uncoupling of SAN cell fate determination and morphogenesis. Single-cell RNA-seq revealed that the presumptive SAN cells of Shox2-/- mutants failed to activate pacemaking program but remained in a progenitor state preceding working myocardium, while both wildtype and dKO SAN cells displayed normal pacemaking cell fate with similar cellular state. Shox2 thus acts as a safeguard but not a determinant to ensure the pacemaking cell fate through the Shox2-Nkx2-5 antagonistic mechanism, which is segregated from its morphogenetic regulatory function in SAN development.
Collapse
Affiliation(s)
- Hua Li
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China.
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA.
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China.
| | - Qinghuang Tang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, 14214, USA
| | - Tianfang Yang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Zhengsen Wang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China
| | - Dainan Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Linyan Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, Sichuan Province, 610021, PR China
| | - Liwen Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Biological Sciences, College of Arts and Sciences, University at Buffalo, Buffalo, NY, 14260, USA
| | - Yaoyi Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Hai Huang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Yanding Zhang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA.
| |
Collapse
|
245
|
Sandor LF, Huh JB, Benko P, Hiraga T, Poliska S, Dobo-Nagy C, Simpson JP, Homer NZM, Mahata B, Gyori DS. De novo steroidogenesis in tumor cells drives bone metastasis and osteoclastogenesis. Cell Rep 2024; 43:113936. [PMID: 38489269 PMCID: PMC10995766 DOI: 10.1016/j.celrep.2024.113936] [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: 07/21/2023] [Revised: 11/08/2023] [Accepted: 02/23/2024] [Indexed: 03/17/2024] Open
Abstract
Osteoclasts play a central role in cancer-cell-induced osteolysis, but the molecular mechanisms of osteoclast activation during bone metastasis formation are incompletely understood. By performing RNA sequencing on a mouse breast carcinoma cell line with higher bone-metastatic potential, here we identify the enzyme CYP11A1 strongly upregulated in osteotropic tumor cells. Genetic deletion of Cyp11a1 in tumor cells leads to a decreased number of bone metastases but does not alter primary tumor growth and lung metastasis formation in mice. The product of CYP11A1 activity, pregnenolone, increases the number and function of mouse and human osteoclasts in vitro but does not alter osteoclast-specific gene expression. Instead, tumor-derived pregnenolone strongly enhances the fusion of pre-osteoclasts via prolyl 4-hydroxylase subunit beta (P4HB), identified as a potential interaction partner of pregnenolone. Taken together, our results demonstrate that Cyp11a1-expressing tumor cells produce pregnenolone, which is capable of promoting bone metastasis formation and osteoclast development via P4HB.
Collapse
Affiliation(s)
- Luca F Sandor
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary
| | - Joon B Huh
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary
| | - Peter Benko
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary
| | - Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, Shiojiri, Nagano 399-0781, Japan
| | - Szilard Poliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Csaba Dobo-Nagy
- Department of Oral Diagnostics, Semmelweis University School of Dentistry, 1088 Budapest, Hungary
| | - Joanna P Simpson
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK; University of Edinburgh/BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Bidesh Mahata
- Department of Pathology, University of Cambridge, Cambridge CB21QP Cambridgeshire, UK
| | - David S Gyori
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary.
| |
Collapse
|
246
|
Chandran RR, Adams TS, Kabir I, Gallardo-Vara E, Kaminski N, Gomperts BN, Greif DM. Dedifferentiated early postnatal lung myofibroblasts redifferentiate in adult disease. Front Cell Dev Biol 2024; 12:1335061. [PMID: 38572485 PMCID: PMC10987733 DOI: 10.3389/fcell.2024.1335061] [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: 11/08/2023] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Alveolarization ensures sufficient lung surface area for gas exchange, and during bulk alveolarization in mice (postnatal day [P] 4.5-14.5), alpha-smooth muscle actin (SMA)+ myofibroblasts accumulate, secrete elastin, and lay down alveolar septum. Herein, we delineate the dynamics of the lineage of early postnatal SMA+ myofibroblasts during and after bulk alveolarization and in response to lung injury. SMA+ lung myofibroblasts first appear at ∼ P2.5 and proliferate robustly. Lineage tracing shows that, at P14.5 and over the next few days, the vast majority of SMA+ myofibroblasts downregulate smooth muscle cell markers and undergo apoptosis. Of note, ∼8% of these dedifferentiated cells and another ∼1% of SMA+ myofibroblasts persist to adulthood. Single cell RNA sequencing analysis of the persistent SMA- cells and SMA+ myofibroblasts in the adult lung reveals distinct gene expression profiles. For instance, dedifferentiated SMA- cells exhibit higher levels of tissue remodeling genes. Most interestingly, these dedifferentiated early postnatal myofibroblasts re-express SMA upon exposure of the adult lung to hypoxia or the pro-fibrotic drug bleomycin. However, unlike during alveolarization, these cells that re-express SMA do not proliferate with hypoxia. In sum, dedifferentiated early postnatal myofibroblasts are a previously undescribed cell type in the adult lung and redifferentiate in response to injury.
Collapse
Affiliation(s)
- Rachana R. Chandran
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Genetics, Yale University School of Medicine, New Haven, CT, United States
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Taylor S. Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Inamul Kabir
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Genetics, Yale University School of Medicine, New Haven, CT, United States
| | - Eunate Gallardo-Vara
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Genetics, Yale University School of Medicine, New Haven, CT, United States
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Brigitte N. Gomperts
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Children’s Discovery and Innovation Institute, Mattel Children’s Hospital, Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
- Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Genetics, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
247
|
Plata-Gómez AB, de Prado-Rivas L, Sanz A, Deleyto-Seldas N, García F, de la Calle Arregui C, Silva C, Caleiras E, Graña-Castro O, Piñeiro-Yáñez E, Krebs J, Leiva-Vega L, Muñoz J, Jain A, Sabio G, Efeyan A. Hepatic nutrient and hormone signaling to mTORC1 instructs the postnatal metabolic zonation of the liver. Nat Commun 2024; 15:1878. [PMID: 38499523 PMCID: PMC10948770 DOI: 10.1038/s41467-024-46032-1] [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: 04/20/2023] [Accepted: 02/09/2024] [Indexed: 03/20/2024] Open
Abstract
The metabolic functions of the liver are spatially organized in a phenomenon called zonation, linked to the differential exposure of portal and central hepatocytes to nutrient-rich blood. The mTORC1 signaling pathway controls cellular metabolism in response to nutrients and insulin fluctuations. Here we show that simultaneous genetic activation of nutrient and hormone signaling to mTORC1 in hepatocytes results in impaired establishment of postnatal metabolic and zonal identity of hepatocytes. Mutant hepatocytes fail to upregulate postnatally the expression of Frizzled receptors 1 and 8, and show reduced Wnt/β-catenin activation. This defect, alongside diminished paracrine Wnt2 ligand expression by endothelial cells, underlies impaired postnatal maturation. Impaired zonation is recapitulated in a model of constant supply of nutrients by parenteral nutrition to piglets. Our work shows the role of hepatocyte sensing of fluctuations in nutrients and hormones for triggering a latent metabolic zonation program.
Collapse
Affiliation(s)
- Ana Belén Plata-Gómez
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Lucía de Prado-Rivas
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Alba Sanz
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Nerea Deleyto-Seldas
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Fernando García
- Proteomics Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Celia de la Calle Arregui
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Camila Silva
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| | - Eduardo Caleiras
- Histopathology Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Osvaldo Graña-Castro
- Bioinformatics Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Department of Basic Medical Sciences, Institute of Applied Molecular Medicine (IMMA-Nemesio Díez), School of Medicine, San Pablo-CEU University, CEU Universities, Boadilla del Monte, Madrid, Spain
| | - Elena Piñeiro-Yáñez
- Bioinformatics Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Joseph Krebs
- Department of Pediatrics, Saint Louis University, Saint Louis, MO, USA
| | - Luis Leiva-Vega
- Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Javier Muñoz
- Proteomics Unit. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Cell Signalling and Clinical Proteomics Group, Biocruces Bizkaia Health Research Institute & Ikerbasque Basque Foundation for Science, Bilbao, Spain
| | - Ajay Jain
- Department of Pediatrics, Saint Louis University, Saint Louis, MO, USA
| | - Guadalupe Sabio
- Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alejo Efeyan
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, Madrid, 28029, Spain.
| |
Collapse
|
248
|
Hansen AH, Hippenmeyer S. Time-lapse imaging of cortical projection neuron migration in mice using mosaic analysis with double markers. STAR Protoc 2024; 5:102795. [PMID: 38165800 PMCID: PMC10797208 DOI: 10.1016/j.xpro.2023.102795] [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: 07/23/2023] [Revised: 10/09/2023] [Accepted: 12/07/2023] [Indexed: 01/04/2024] Open
Abstract
Mosaic analysis with double markers (MADM) technology enables the sparse labeling of genetically defined neurons. We present a protocol for time-lapse imaging of cortical projection neuron migration in mice using MADM. We describe steps for the isolation, culturing, and 4D imaging of neuronal dynamics in MADM-labeled brain tissue. While this protocol is compatible with other single-cell labeling methods, the MADM approach provides a genetic platform for the functional assessment of cell-autonomous candidate gene function and the relative contribution of non-cell-autonomous effects. For complete details on the use and execution of this protocol, please refer to Hansen et al. (2022),1 Contreras et al. (2021),2 and Amberg and Hippenmeyer (2021).3.
Collapse
Affiliation(s)
- Andi H Hansen
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria.
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
249
|
Moore E, Zhao R, McKinney MC, Yi K, Wood C, Trainor P. Cell extrusion - a novel mechanism driving neural crest cell delamination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.09.584232. [PMID: 38559094 PMCID: PMC10979875 DOI: 10.1101/2024.03.09.584232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Neural crest cells (NCC) comprise a heterogeneous population of cells with variable potency, that contribute to nearly every tissue and organ system throughout the body. Considered unique to vertebrates, NCC are transiently generated within the dorsolateral region of the neural plate or neural tube, during neurulation. Their delamination and migration are crucial events in embryo development as the differentiation of NCC is heavily influenced by their final resting locations. Previous work in avian and aquatic species has shown that NCC delaminate via an epithelial-mesenchymal transition (EMT), which transforms these stem and progenitor cells from static polarized epithelial cells into migratory mesenchymal cells with fluid front and back polarity. However, the cellular and molecular drivers facilitating NCC delamination in mammals are poorly understood. We performed live timelapse imaging of NCC delamination in mouse embryos and discovered a group of cells that exit the neuroepithelium as isolated round cells, which then halt for a short period prior to acquiring the mesenchymal migratory morphology classically associated with most delaminating NCC. High magnification imaging and protein localization analyses of the cytoskeleton, together with measurements of pressure and tension of delaminating NCC and neighboring neuroepithelial cells, revealed these round NCC are extruded from the neuroepithelium prior to completion of EMT. Furthermore, we demonstrate that cranial NCC are extruded through activation of the mechanosensitive ion channel, PIEZO1, a key regulator of the live cell extrusion pathway, revealing a new role for PIEZO1 in neural crest cell development. Our results elucidating the cellular and molecular dynamics orchestrating NCC delamination support a model in which high pressure and tension in the neuroepithelium results in activation of the live cell extrusion pathway and delamination of a subpopulation of NCC in parallel with EMT. This model has broad implications for our understanding of cell delamination in development and disease.
Collapse
Affiliation(s)
- Emma Moore
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mary C McKinney
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Paul Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
250
|
Banik J, Moreira ARS, Lim J, Tomlinson S, Hardy LL, Lagasse A, Haney A, Crimmins MR, Boehm U, Odle AK, MacNicol MC, Childs GV, MacNicol AM. The Musashi RNA binding proteins direct the translational activation of key pituitary mRNAs. Sci Rep 2024; 14:5918. [PMID: 38467682 PMCID: PMC10928108 DOI: 10.1038/s41598-024-56002-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/29/2024] [Indexed: 03/13/2024] Open
Abstract
The pituitary functions as a master endocrine gland that secretes hormones critical for regulation of a wide variety of physiological processes including reproduction, growth, metabolism and stress responses. The distinct hormone-producing cell lineages within the pituitary display remarkable levels of cell plasticity that allow remodeling of the relative proportions of each hormone-producing cell population to meet organismal demands. The molecular mechanisms governing pituitary cell plasticity have not been fully elucidated. Our recent studies have implicated a role for the Musashi family of sequence-specific mRNA binding proteins in the control of pituitary hormone production, pituitary responses to hypothalamic stimulation and modulation of pituitary transcription factor expression in response to leptin signaling. To date, these actions of Musashi in the pituitary appear to be mediated through translational repression of the target mRNAs. Here, we report Musashi1 directs the translational activation, rather than repression, of the Prop1, Gata2 and Nr5a1 mRNAs which encode key pituitary lineage specification factors. We observe that Musashi1 further directs the translational activation of the mRNA encoding the glycolipid Neuronatin (Nnat) as determined both in mRNA reporter assays as well as in vivo. Our findings suggest a complex bifunctional role for Musashi1 in the control of pituitary cell function.
Collapse
Affiliation(s)
- Jewel Banik
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Ana Rita Silva Moreira
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Juchan Lim
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Sophia Tomlinson
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Linda L Hardy
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Alex Lagasse
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Anessa Haney
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Meghan R Crimmins
- Arkansas Children's Nutrition Center, Arkansas Children's Hospital, Little Rock, AR, USA
| | - Ulrich Boehm
- Department of Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg, Germany
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA.
| |
Collapse
|