151
|
Krause W, King D, Horsley V. Transcriptional analysis of efferocytosis in mouse skin wounds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607219. [PMID: 39185146 PMCID: PMC11343138 DOI: 10.1101/2024.08.12.607219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Defects in apoptotic cell clearance, or efferocytosis, can cause inflammatory diseases and prevent tissue repair due in part to inducing a pro-repair transcriptional program in phagocytic cells like macrophages. While the cellular machinery and metabolic pathways involved in efferocytosis have been characterized, the precise efferocytic response of macrophages is dependent on the identity and macromolecular cues of apoptotic cells, and the complex tissue microenvironment in which efferocytosis occurs. Here, we find that macrophages undergoing active efferocytosis in mid-stage mouse skin wounds in vivo display a pro-repair gene program, while efferocytosis of apoptotic skin fibroblasts in vitro also induces an inflammatory transcription response. These data provide a resource for understanding how the skin wound environment influences macrophage efferocytosis and will be useful for future investigations that define the role of efferocytosis during tissue repair.
Collapse
Affiliation(s)
- Will Krause
- Dept. of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Diane King
- SunnyCrest Bioinformatics, Flemington, New Jersey, USA
| | - Valerie Horsley
- Dept. of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut, USA
- Dept. of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
152
|
Gao X, Carpenter RS, Boulais PE, Zhang D, Marlein CR, Li H, Smith M, Chung DJ, Maryanovich M, Will B, Steidl U, Frenette PS. Regulation of the hematopoietic stem cell pool by C-Kit-associated trogocytosis. Science 2024; 385:eadp2065. [PMID: 39116219 PMCID: PMC11533977 DOI: 10.1126/science.adp2065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/14/2024] [Indexed: 08/10/2024]
Abstract
Hematopoietic stem cells (HSCs) are routinely mobilized from the bone marrow (BM) to the blood circulation for clinical transplantation. However, the precise mechanisms by which individual stem cells exit the marrow are not understood. This study identified cell-extrinsic and molecular determinants of a mobilizable pool of blood-forming stem cells. We found that a subset of HSCs displays macrophage-associated markers on their cell surface. Although fully functional, these HSCs are selectively niche-retained as opposed to stem cells lacking macrophage markers, which exit the BM upon forced mobilization. Macrophage markers on HSCs could be acquired through direct transfer by trogocytosis, regulated by receptor tyrosine-protein kinase C-Kit (CD117), from BM-resident macrophages in mouse and human settings. Our study provides proof of concept that adult stem cells utilize trogocytosis to rapidly establish and activate function-modulating molecular mechanisms.
Collapse
Affiliation(s)
- Xin Gao
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Wisconsin Blood Cancer Research Institute, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Randall S. Carpenter
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Philip E. Boulais
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dachuan Zhang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Christopher R. Marlein
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Huihui Li
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Matthew Smith
- Wisconsin Blood Cancer Research Institute, Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - David J. Chung
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Maryanovich
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine–Montefiore Health System, Bronx, NY, USA
| | - Britta Will
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine–Montefiore Health System, Bronx, NY, USA
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ulrich Steidl
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine–Montefiore Health System, Bronx, NY, USA
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Paul S. Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
153
|
Zheng H, Zhao P, Tan Z, Yu WM, Werner J, Stieglitz E, Porter C, Chandrakasan S, Wechsler D, Mendez-Ferrer S, Qu CK. Prototypical innate immune mechanism hijacked by leukemia-initiating mutant stem cells for selective advantage and immune evasion in Ptpn11-associated juvenile myelomonocytic leukemia. RESEARCH SQUARE 2024:rs.3.rs-4450642. [PMID: 39149498 PMCID: PMC11326406 DOI: 10.21203/rs.3.rs-4450642/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Juvenile myelomonocytic leukemia (JMML), a clonal hematologic malignancy, originates from mutated hematopoietic stem cells (HSCs). The mechanism sustaining the persistence of mutant stem cells, leading to leukemia development, remains elusive. In this study, we conducted comprehensive examination of gene expression profiles, transcriptional factor regulons, and cell compositions/interactions throughout various stages of tumor cell development in Ptpn11 mutation-associated JMML. Our analyses revealed that leukemia-initiating Ptpn11 E76K/+ mutant stem cells exhibited de novo activation of the myeloid transcriptional program and aberrant developmental trajectories. These mutant stem cells displayed significantly elevated expression of innate immunity-associated anti-microbial peptides and pro-inflammatory proteins, particularly S100a9 and S100a8. Biological experiments confirmed that S100a9/S100a8 conferred a selective advantage to the leukemia-initiating cells through autocrine effects and facilitated immune evasion by recruiting and promoting immune suppressive myeloid-derived suppressor cells (MDSCs) in the microenvironment. Importantly, pharmacological inhibition of S100a9/S100a8 signaling effectively impeded leukemia development from Ptpn11 E76K/+ mutant stem cells. These findings collectively suggest that JMML tumor-initiating cells exploit evolutionarily conserved innate immune and inflammatory mechanisms to establish clonal dominance.
Collapse
Affiliation(s)
- Hong Zheng
- Department of Pediatrics, Aflac Cancer & Blood Disorders Center, Winship Cancer Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, USA; Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Peng Zhao
- Department of Pediatrics, Aflac Cancer & Blood Disorders Center, Winship Cancer Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, USA
| | - Zhenya Tan
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Wen-Mei Yu
- Department of Pediatrics, Aflac Cancer & Blood Disorders Center, Winship Cancer Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, USA
| | - Juwita Werner
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of California San Francisco, San Francisco, USA
| | - Elliot Stieglitz
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of California San Francisco, San Francisco, USA
| | - Chris Porter
- Department of Pediatrics, Aflac Cancer & Blood Disorders Center, Winship Cancer Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, USA
| | - Shanmuganathan Chandrakasan
- Department of Pediatrics, Aflac Cancer & Blood Disorders Center, Winship Cancer Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, USA
| | - Daniel Wechsler
- Department of Pediatrics, Aflac Cancer & Blood Disorders Center, Winship Cancer Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, USA
| | - Simon Mendez-Ferrer
- Department of Hematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Cheng-Kui Qu
- Department of Pediatrics, Aflac Cancer & Blood Disorders Center, Winship Cancer Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, USA
| |
Collapse
|
154
|
Metwally E, Sanchez Solano A, Lavanderos B, Yamasaki E, Thakore P, McClenaghan C, Rios N, Radi R, Feng Earley Y, Nichols CG, Earley S. Mitochondrial Ca2+-coupled generation of reactive oxygen species, peroxynitrite formation, and endothelial dysfunction in Cantú syndrome. JCI Insight 2024; 9:e176212. [PMID: 39088268 PMCID: PMC11385080 DOI: 10.1172/jci.insight.176212] [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: 09/27/2023] [Accepted: 07/25/2024] [Indexed: 08/03/2024] Open
Abstract
Cantú syndrome is a multisystem disorder caused by gain-of-function (GOF) mutations in KCNJ8 and ABCC9, the genes encoding the pore-forming inward rectifier Kir6.1 and regulatory sulfonylurea receptor SUR2B subunits, respectively, of vascular ATP-sensitive K+ (KATP) channels. In this study, we investigated changes in the vascular endothelium in mice in which Cantú syndrome-associated Kcnj8 or Abcc9 mutations were knocked in to the endogenous loci. We found that endothelium-dependent dilation was impaired in small mesenteric arteries from Cantú mice. Loss of endothelium-dependent vasodilation led to increased vasoconstriction in response to intraluminal pressure or treatment with the adrenergic receptor agonist phenylephrine. We also found that either KATP GOF or acute activation of KATP channels with pinacidil increased the amplitude and frequency of wave-like Ca2+ events generated in the endothelium in response to the vasodilator agonist carbachol. Increased cytosolic Ca2+ signaling activity in arterial endothelial cells from Cantú mice was associated with elevated mitochondrial [Ca2+] and enhanced reactive oxygen species (ROS) and peroxynitrite levels. Scavenging intracellular or mitochondrial ROS restored endothelium-dependent vasodilation in the arteries of mice with KATP GOF mutations. We conclude that mitochondrial Ca2+ overload and ROS generation, which subsequently leads to nitric oxide consumption and peroxynitrite formation, cause endothelial dysfunction in mice with Cantú syndrome.
Collapse
Affiliation(s)
- Elsayed Metwally
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Alfredo Sanchez Solano
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Boris Lavanderos
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Evan Yamasaki
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Conor McClenaghan
- Departments of Pharmacology and Medicine, Center for Advanced Biotechnology and Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Natalia Rios
- Departamento de Bioquímica, Facultad de Medicina, and
- Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, and
- Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Yumei Feng Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Colin G. Nichols
- Center for the Investigation of Membrane Excitability Diseases and Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| |
Collapse
|
155
|
Menche C, Schuhwerk H, Armstark I, Gupta P, Fuchs K, van Roey R, Mosa MH, Hartebrodt A, Hajjaj Y, Clavel Ezquerra A, Selvaraju MK, Geppert CI, Bärthel S, Saur D, Greten FR, Brabletz S, Blumenthal DB, Weigert A, Brabletz T, Farin HF, Stemmler MP. ZEB1-mediated fibroblast polarization controls inflammation and sensitivity to immunotherapy in colorectal cancer. EMBO Rep 2024; 25:3406-3431. [PMID: 38937629 PMCID: PMC11315988 DOI: 10.1038/s44319-024-00186-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024] Open
Abstract
The EMT-transcription factor ZEB1 is heterogeneously expressed in tumor cells and in cancer-associated fibroblasts (CAFs) in colorectal cancer (CRC). While ZEB1 in tumor cells regulates metastasis and therapy resistance, its role in CAFs is largely unknown. Combining fibroblast-specific Zeb1 deletion with immunocompetent mouse models of CRC, we observe that inflammation-driven tumorigenesis is accelerated, whereas invasion and metastasis in sporadic cancers are reduced. Single-cell transcriptomics, histological characterization, and in vitro modeling reveal a crucial role of ZEB1 in CAF polarization, promoting myofibroblastic features by restricting inflammatory activation. Zeb1 deficiency impairs collagen deposition and CAF barrier function but increases NFκB-mediated cytokine production, jointly promoting lymphocyte recruitment and immune checkpoint activation. Strikingly, the Zeb1-deficient CAF repertoire sensitizes to immune checkpoint inhibition, offering a therapeutic opportunity of targeting ZEB1 in CAFs and its usage as a prognostic biomarker. Collectively, we demonstrate that ZEB1-dependent plasticity of CAFs suppresses anti-tumor immunity and promotes metastasis.
Collapse
Affiliation(s)
- Constantin Menche
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Harald Schuhwerk
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Isabell Armstark
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Pooja Gupta
- Core Unit for Bioinformatics, Data Integration and Analysis, Center for Medical Information and Communication Technology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Kathrin Fuchs
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Ruthger van Roey
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Mohammed H Mosa
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Anne Hartebrodt
- Biomedical Network Science Lab, Department Artificial Intelligence in Biomedical Engineering (AIBE), FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Yussuf Hajjaj
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Ana Clavel Ezquerra
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Manoj K Selvaraju
- Core Unit for Bioinformatics, Data Integration and Analysis, Center for Medical Information and Communication Technology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Carol I Geppert
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Stefanie Bärthel
- Division of Translational Cancer Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, Klinikum rechts der Isar, School of Medicine, Technische Universität München, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Dieter Saur
- Division of Translational Cancer Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, Klinikum rechts der Isar, School of Medicine, Technische Universität München, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Florian R Greten
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
- German Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - David B Blumenthal
- Biomedical Network Science Lab, Department Artificial Intelligence in Biomedical Engineering (AIBE), FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Weigert
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- Institute of Biochemistry I, Goethe University, Frankfurt am Main, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU Erlangen-Nürnberg, Erlangen, Germany.
| | - Henner F Farin
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany.
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany.
- German Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
156
|
Satta JP, Lindström R, Myllymäki SM, Lan Q, Trela E, Prunskaite-Hyyryläinen R, Kaczyńska B, Voutilainen M, Kuure S, Vainio SJ, Mikkola ML. Exploring the principles of embryonic mammary gland branching morphogenesis. Development 2024; 151:dev202179. [PMID: 39092607 DOI: 10.1242/dev.202179] [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: 07/13/2023] [Accepted: 06/21/2024] [Indexed: 08/04/2024]
Abstract
Branching morphogenesis is a characteristic feature of many essential organs, such as the lung and kidney, and most glands, and is the net result of two tissue behaviors: branch point initiation and elongation. Each branched organ has a distinct architecture customized to its physiological function, but how patterning occurs in these ramified tubular structures is a fundamental problem of development. Here, we use quantitative 3D morphometrics, time-lapse imaging, manipulation of ex vivo cultured mouse embryonic organs and mice deficient in the planar cell polarity component Vangl2 to address this question in the developing mammary gland. Our results show that the embryonic epithelial trees are highly complex in topology owing to the flexible use of two distinct modes of branch point initiation: lateral branching and tip bifurcation. This non-stereotypy was contrasted by the remarkably constant average branch frequency, indicating a ductal growth invariant, yet stochastic, propensity to branch. The probability of branching was malleable and could be tuned by manipulating the Fgf10 and Tgfβ1 pathways. Finally, our in vivo data and ex vivo time-lapse imaging suggest the involvement of tissue rearrangements in mammary branch elongation.
Collapse
Affiliation(s)
- Jyoti P Satta
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Riitta Lindström
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Satu-Marja Myllymäki
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Qiang Lan
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Ewelina Trela
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | | | - Beata Kaczyńska
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Maria Voutilainen
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Satu Kuure
- GM-unit, Laboratory Animal Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Seppo J Vainio
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu 90014, Finland
- Kvantum Institute, Infotech Oulu, University of Oulu, Oulu 90014, Finland
| | - Marja L Mikkola
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| |
Collapse
|
157
|
Luo W, Fu X, Huang H, Wu P, Wang Y, Liu Z, He S, Pang L, Ren D, Cui Y. Planar Cell Polarity in the Multiciliated Epithelial Lining of the Mouse Eustachian Tube. Laryngoscope 2024; 134:3795-3801. [PMID: 38613460 DOI: 10.1002/lary.31451] [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: 11/30/2023] [Revised: 03/26/2024] [Accepted: 04/02/2024] [Indexed: 04/15/2024]
Abstract
OBJECTIVES Planar cell polarity (PCP) signaling, essential for uniform alignment and directional beating of motile cilia, has been investigated in multiciliated epithelia. As a complex structure connecting the middle ear to the nasopharynx, the eustachian tube (ET) is important in the onset of ear-nose-throat diseases. However, PCP signaling, including the orientation that is important for ciliary motility and clearance function in the ET, has not been studied. We evaluated PCP in the ET epithelium. STUDY DESIGN Morphometric examination of the mouse ET. METHODS We performed electron microscopy to assess ciliary polarity in the mouse ET, along with immunohistochemical analysis of PCP protein localization in the ET epithelium. RESULTS We discovered PCP in the ET epithelium. Motile cilia were aligned in the same direction in individual and neighboring cells; this alignment manifested as ciliary polarity in multiciliated cells. Additionally, PCP proteins were asymmetrically localized between adjacent cells in the plane of the ET. CONCLUSIONS The multiciliated ET epithelium exhibits polarization, suggesting novel structural features that may be critical for ET function. LEVEL OF EVIDENCE NA Laryngoscope, 134:3795-3801, 2024.
Collapse
Affiliation(s)
- Wenwei Luo
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiao Fu
- Department of Otolaryngology-Head and Neck Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Hongming Huang
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Peina Wu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yanmei Wang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhifeng Liu
- Department of Otolaryngology, Longgang E.N.T hospital & Institute of E.N.T, Shenzhen, China
| | - Shiqi He
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Limin Pang
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Dongdong Ren
- Department of Otolaryngology-Head and Neck Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Yong Cui
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
158
|
Vaidya A, Moore S, Chatterjee S, Guerrero E, Kim M, Farbiak L, Dilliard SA, Siegwart DJ. Expanding RNAi to Kidneys, Lungs, and Spleen via Selective ORgan Targeting (SORT) siRNA Lipid Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313791. [PMID: 38973655 DOI: 10.1002/adma.202313791] [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: 12/17/2023] [Revised: 06/14/2024] [Indexed: 07/09/2024]
Abstract
Inhibition of disease-causing mutations using RNA interference (RNAi) has resulted in clinically approved medicines with additional candidates in late stage trials. However, targetable tissues currently in preclinical development are limited to liver following systemic intravenous (IV) administration because predictable delivery of siRNA to non-liver tissues remains an unsolved challenge. Here, evidence of durable extrahepatic gene silencing enabled by siRNA Selective ORgan Targeting lipid nanoparticles (siRNA SORT LNPs) to the kidneys, lungs, and spleen is provided. LNPs excel at dose-dependent silencing of tissue-enriched endogenous targets resulting in 60%-80% maximal knockdown after a single IV injection and up to 88% downregulation of protein expression in mouse lungs after two doses. To examine knockdown potency and unbiased organ targeting, B6.129TdTom/EGFP mice that constitutively express the TdTomato transgene across all cell types are utilized to demonstrate 58%, 45%, and 15% reduction in TdTomato fluorescence in lungs, spleen, and kidneys, respectively. Finally, physiological relevance of siRNA SORT LNP-mediated gene silencing is established via acute suppression of endogenous Tie2 which induces lung-specific phenotypic alteration of vascular endothelial barrier. Due to plethora of extrahepatic diseases that may benefit from RNAi interventions, it is anticipated that the findings will expand preclinical landscape of therapeutic targets beyond the liver.
Collapse
Affiliation(s)
- Amogh Vaidya
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Stephen Moore
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Sumanta Chatterjee
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Erick Guerrero
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Minjeong Kim
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Lukas Farbiak
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Sean A Dilliard
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Daniel J Siegwart
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| |
Collapse
|
159
|
Bywaters BC, Trache A, Rivera GM. Modulation of arterial intima stiffness by disturbed blood flow. Exp Biol Med (Maywood) 2024; 249:10090. [PMID: 39143955 PMCID: PMC11323813 DOI: 10.3389/ebm.2024.10090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/09/2024] [Indexed: 08/16/2024] Open
Abstract
The intima, comprising the endothelium and the subendothelial matrix, plays a crucial role in atherosclerosis pathogenesis. The mechanical stress arising from disturbed blood flow (d-flow) and the stiffening of the arterial wall contributes to endothelial dysfunction. However, the specific impacts of these physical forces on the mechanical environment of the intima remain undetermined. Here, we investigated whether inhibiting collagen crosslinking could ameliorate the detrimental effects of persistent d-flow on the mechanical properties of the intima. Partial ligation of the left carotid artery (LCA) was performed in C57BL/6J mice, inducing d-flow. The right carotid artery (RCA) served as an internal control. Carotids were collected 2 days and 2 weeks after surgery to study acute and chronic effects of d-flow on the mechanical phenotype of the intima. The chronic effects of d-flow were decoupled from the ensuing arterial wall stiffening by administration of β-aminopropionitrile (BAPN), an inhibitor of collagen crosslinking by lysyl oxidase (LOX) enzymes. Atomic force microscopy (AFM) was used to determine stiffness of the endothelium and the denuded subendothelial matrix in en face carotid preparations. The stiffness of human aortic endothelial cells (HAEC) cultured on soft and stiff hydrogels was also determined. Acute exposure to d-flow caused a slight decrease in endothelial stiffness in male mice but had no effect on the stiffness of the subendothelial matrix in either sex. Regardless of sex, the intact endothelium was softer than the subendothelial matrix. In contrast, exposure to chronic d-flow led to a substantial increase in the endothelial and subendothelial stiffness in both sexes. The effects of chronic d-flow were largely prevented by concurrent BAPN administration. In addition, HAEC displayed reduced stiffness when cultured on soft vs. stiff hydrogels. We conclude that chronic d-flow results in marked stiffening of the arterial intima, which can be effectively prevented by inhibition of collagen crosslinking.
Collapse
Affiliation(s)
- Briana C. Bywaters
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Andreea Trache
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, TX, United States
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Gonzalo M. Rivera
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
160
|
Sivaraj KK, Majev PG, Dharmalingam B, Schröder S, Banjanin B, Stehling M, Zeuschner D, Nordheim A, Schneider RK, Adams RH. Endothelial LATS2 is a suppressor of bone marrow fibrosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:951-969. [PMID: 39155965 PMCID: PMC11324521 DOI: 10.1038/s44161-024-00508-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/13/2024] [Indexed: 08/20/2024]
Abstract
Myelofibrosis and osteosclerosis are fibrotic diseases disrupting bone marrow function that occur in various leukemias but also in response to non-malignant alterations in hematopoietic cells. Here we show that endothelial cell-specific inactivation of the Lats2 gene, encoding Hippo kinase large tumor suppressor kinase 2, or overexpression of the downstream effector YAP1 induce myofibroblast formation and lead to extensive fibrosis and osteosclerosis, which impair bone marrow function and cause extramedullary hematopoiesis in the spleen. Mechanistically, loss of LATS2 induces endothelial-to-mesenchymal transition, resulting in increased expression of extracellular matrix and secreted signaling molecules. Changes in endothelial cells involve increased expression of serum response factor target genes, and, strikingly, major aspects of the LATS2 mutant phenotype are rescued by inactivation of the Srf gene. These findings identify the endothelium as a driver of bone marrow fibrosis, which improves understanding of myelofibrotic and osteosclerotic diseases, for which drug therapies are currently lacking.
Collapse
Affiliation(s)
- Kishor K. Sivaraj
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Paul-Georg Majev
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | | | - Silke Schröder
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Bella Banjanin
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Martin Stehling
- Flow Cytometry Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dagmar Zeuschner
- Electron Microscopy Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Alfred Nordheim
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
- Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| | - Rebekka K. Schneider
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Oncode Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
- Institute for Cell and Tumor Biology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Ralf H. Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
161
|
Perrin S, Ethel M, Bretegnier V, Goachet C, Wotawa CA, Luka M, Coulpier F, Masson C, Ménager M, Colnot C. Single nuclei transcriptomics reveal the differentiation trajectories of periosteal skeletal/stem progenitor cells in bone regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.23.546220. [PMID: 39211065 PMCID: PMC11361009 DOI: 10.1101/2023.06.23.546220] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Bone regeneration is mediated by skeletal stem/progenitor cells (SSPCs) that are mainly recruited from the periosteum after bone injury. The composition of the periosteum and the steps of SSPC activation and differentiation remain poorly understood. Here, we generated a single-nuclei atlas of the periosteum at steady-state and of the fracture site during early stages of bone repair ( https://fracture-repair-atlas.cells.ucsc.edu ). We identified periosteal SSPCs expressing stemness markers ( Pi16 and Ly6a /SCA1) and responding to fracture by adopting an injury-induced fibrogenic cell (IIFC) fate, prior to undergoing osteogenesis or chondrogenesis. We identified distinct gene cores associated with IIFCs and their engagement into osteogenesis and chondrogenesis involving Notch, Wnt and the circadian clock signaling respectively. Finally, we show that IIFCs are the main source of paracrine signals in the fracture environment, suggesting a crucial paracrine role of this transient IIFC population during fracture healing. Overall, our study provides a complete temporal topography of the early stages of fracture healing and the dynamic response of periosteal SSPCs to injury, redefining our knowledge of bone regeneration.
Collapse
|
162
|
Xu Y, Ma S, Cui H, Chen J, Xu S, Gong F, Golubovic A, Zhou M, Wang KC, Varley A, Lu RXZ, Wang B, Li B. AGILE platform: a deep learning powered approach to accelerate LNP development for mRNA delivery. Nat Commun 2024; 15:6305. [PMID: 39060305 PMCID: PMC11282250 DOI: 10.1038/s41467-024-50619-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Ionizable lipid nanoparticles (LNPs) are seeing widespread use in mRNA delivery, notably in SARS-CoV-2 mRNA vaccines. However, the expansion of mRNA therapies beyond COVID-19 is impeded by the absence of LNPs tailored for diverse cell types. In this study, we present the AI-Guided Ionizable Lipid Engineering (AGILE) platform, a synergistic combination of deep learning and combinatorial chemistry. AGILE streamlines ionizable lipid development with efficient library design, in silico lipid screening via deep neural networks, and adaptability to diverse cell lines. Using AGILE, we rapidly design, synthesize, and evaluate ionizable lipids for mRNA delivery, selecting from a vast library. Intriguingly, AGILE reveals cell-specific preferences for ionizable lipids, indicating tailoring for optimal delivery to varying cell types. These highlight AGILE's potential in expediting the development of customized LNPs, addressing the complex needs of mRNA delivery in clinical practice, thereby broadening the scope and efficacy of mRNA therapies.
Collapse
Affiliation(s)
- Yue Xu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Shihao Ma
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Vector Institute for Artificial Intelligence, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Haotian Cui
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Vector Institute for Artificial Intelligence, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Jingan Chen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Shufen Xu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Fanglin Gong
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Alex Golubovic
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Muye Zhou
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Kevin Chang Wang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Andrew Varley
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Rick Xing Ze Lu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Bo Wang
- Department of Computer Science, University of Toronto, Toronto, ON, Canada.
- Vector Institute for Artificial Intelligence, Toronto, ON, Canada.
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada.
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada.
| | - Bowen Li
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
- Department of Chemistry, University of Toronto, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
163
|
Park H, Song J, Jeong HW, Grönloh MLB, Koh BI, Bovay E, Kim KP, Klotz L, Thistlethwaite PA, van Buul JD, Sorokin L, Adams RH. Apelin modulates inflammation and leukocyte recruitment in experimental autoimmune encephalomyelitis. Nat Commun 2024; 15:6282. [PMID: 39060233 PMCID: PMC11282314 DOI: 10.1038/s41467-024-50540-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Demyelination due to autoreactive T cells and inflammation in the central nervous system are principal features of multiple sclerosis (MS), a chronic and highly disabling human disease affecting brain and spinal cord. Here, we show that treatment with apelin, a secreted peptide ligand for the G protein-coupled receptor APJ/Aplnr, is protective in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Apelin reduces immune cell entry into the brain, delays the onset and reduces the severity of EAE. Apelin affects the trafficking of leukocytes through the lung by modulating the expression of cell adhesion molecules that mediate leukocyte recruitment. In addition, apelin induces the internalization and desensitization of its receptor in endothelial cells (ECs). Accordingly, protection against EAE major outcomes of apelin treatment are phenocopied by loss of APJ/Aplnr function, achieved by EC-specific gene inactivation in mice or knockdown experiments in cultured primary endothelial cells. Our findings highlight the importance of the lung-brain axis in neuroinflammation and indicate that apelin targets the transendothelial migration of immune cells into the lung during acute inflammation.
Collapse
Affiliation(s)
- Hongryeol Park
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany.
| | - Jian Song
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Interfaculty Centre (CIMIC), University of Münster, Münster, Germany
| | - Hyun-Woo Jeong
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Max L B Grönloh
- Vascular Cell Biology Lab, Department of Medical Biochemistry, Amsterdam UMC, and Section Molecular Cytology at Swammerdam Institute for Life Sciences, Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands
| | - Bong Ihn Koh
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Esther Bovay
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Kee-Pyo Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Luisa Klotz
- Department of Neurology, University of Münster, Münster, Germany
| | | | - Jaap D van Buul
- Vascular Cell Biology Lab, Department of Medical Biochemistry, Amsterdam UMC, and Section Molecular Cytology at Swammerdam Institute for Life Sciences, Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Interfaculty Centre (CIMIC), University of Münster, Münster, Germany
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany.
| |
Collapse
|
164
|
Linneberg-Agerholm M, Sell AC, Redó-Riveiro A, Perera M, Proks M, Knudsen TE, Barral A, Manzanares M, Brickman JM. The primitive endoderm supports lineage plasticity to enable regulative development. Cell 2024; 187:4010-4029.e16. [PMID: 38917790 PMCID: PMC11290322 DOI: 10.1016/j.cell.2024.05.051] [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: 05/19/2023] [Revised: 02/27/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024]
Abstract
Mammalian blastocyst formation involves the specification of the trophectoderm followed by the differentiation of the inner cell mass into embryonic epiblast and extra-embryonic primitive endoderm (PrE). During this time, the embryo maintains a window of plasticity and can redirect its cellular fate when challenged experimentally. In this context, we found that the PrE alone was sufficient to regenerate a complete blastocyst and continue post-implantation development. We identify an in vitro population similar to the early PrE in vivo that exhibits the same embryonic and extra-embryonic potency and can form complete stem cell-based embryo models, termed blastoids. Commitment in the PrE is suppressed by JAK/STAT signaling, collaborating with OCT4 and the sustained expression of a subset of pluripotency-related transcription factors that safeguard an enhancer landscape permissive for multi-lineage differentiation. Our observations support the notion that transcription factor persistence underlies plasticity in regulative development and highlight the importance of the PrE in perturbed development.
Collapse
Affiliation(s)
- Madeleine Linneberg-Agerholm
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Annika Charlotte Sell
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Alba Redó-Riveiro
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Marta Perera
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Martin Proks
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Teresa E Knudsen
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Antonio Barral
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, 28049 Madrid, Spain
| | - Miguel Manzanares
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, 28049 Madrid, Spain
| | - Joshua M Brickman
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, 2200 Copenhagen N, Denmark.
| |
Collapse
|
165
|
Fraissenon A, Bayard C, Morin G, Benichi S, Hoguin C, Protic S, Zerbib L, Ladraa S, Firpion M, Blauwblomme T, Naggara O, Duruisseaux M, Delous M, Boitel C, Bringuier PP, Payen L, Legendre C, Kaltenbach S, Balducci E, Villarese P, Asnafi V, Bisdorff A, Guibaud L, Canaud G. Sotorasib for Vascular Malformations Associated with KRAS G12C Mutation. N Engl J Med 2024; 391:334-342. [PMID: 39018528 DOI: 10.1056/nejmoa2309160] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
KRAS gain-of-function mutations are frequently observed in sporadic arteriovenous malformations. The mechanisms underlying the progression of such KRAS-driven malformations are still incompletely understood, and no treatments for the condition are approved. Here, we show the effectiveness of sotorasib, a specific KRAS G12C inhibitor, in reducing the volume of vascular malformations and improving survival in two mouse models carrying a mosaic Kras G12C mutation. We then administered sotorasib to two adult patients with severe KRAS G12C-related arteriovenous malformations. Both patients had rapid reductions in symptoms and arteriovenous malformation size. Targeting KRAS G12C appears to be a promising therapeutic approach for patients with KRAS G12C-related vascular malformations. (Funded by the European Research Council and others.).
Collapse
Affiliation(s)
- Antoine Fraissenon
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Charles Bayard
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Gabriel Morin
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Sandro Benichi
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Clément Hoguin
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Sanela Protic
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Lola Zerbib
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Sophia Ladraa
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Marina Firpion
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Thomas Blauwblomme
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Olivier Naggara
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Michael Duruisseaux
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Marion Delous
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Clothilde Boitel
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Pierre-Paul Bringuier
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Léa Payen
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Christophe Legendre
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Sophie Kaltenbach
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Estelle Balducci
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Patrick Villarese
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Vahid Asnafi
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Annouk Bisdorff
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Laurent Guibaud
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| | - Guillaume Canaud
- From Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron (A.F., L.G.), CREATIS Unité Mixte de Recherche 5220, Villeurbanne (A.F.), INSERM Unité 1151, Institut Necker-Enfants Malades (A.F., C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., V.A., L.G., G.C.), Université Paris Cité (C. Bayard, G.M., S.B., C.H., S.P., L.Z., S.L., M.F., T.B., O.N., C.L., E.B., V.A., L.G., G.C.), Unité de Médecine Translationnelle et Thérapies Ciblées (C. Bayard, G.M., C.H., S.P., L.Z., S.L., M.F., L.G., G.C.), Service de Neurochirurgie Pédiatrique (S.B., T.B.), Service de Néphrologie et Transplantation Adultes (C.L.), and Laboratoire d'Oncohématologie (S.K., E.B., P.V., V.A.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Département de Neuroradiologie, Hôpital Lariboisière, AP-HP (A.B.), and Service de Neuroradiologie Interventionnelle, Hôpital Sainte Anne, AP-HP (O.N.), Paris, Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne (A.F.), the Respiratory Department and Early phase EPSILYON Est, Louis Pradel Hospital, Oncopharmacology Laboratory, Cancer Research Center of Lyon, Unité Mixte de Recherche INSERM 1052, Center National de la Recherche Scientifique (CNRS) 5286 (M. Duruisseaux), Centre de Recherche en Neurosciences de Lyon, INSERM Unité 1028, CNRS Unité Mixte de Recherche 5292 (M. Delous, C. Boitel), and the Institute of Pharmaceutical and Biological Sciences (L.P.), Université Claude Bernard Lyon 1, and Service d'Anatomie Pathologique, Hôpital Edouard Herriot, Hospices Civils de Lyon (P.-P.B.), Lyon, the Circulating Cancer Program, Cancer Institute (L.P.), and Laboratoire de Biologie Médicale Multi Sites du Centre Hospitalier Universitaire de Lyon, Service de Biochimie et Biologie Moléculaire (L.P.), Hospices Civils de Lyon, and the Center for Innovation in Cancerology of Lyon, EA 3738, Faculty of Medicine and Maieutic Lyon Sud, Université Claude Bernard Lyon 1 (L.P.), Oullins-Pierre-Bénite - all in France
| |
Collapse
|
166
|
Bunn KE, Giese-Byrne BG, Pua HH. Th2 cell extracellular vesicles promote eosinophil survival through the cytokine cargo IL-3 and prolong airway eosinophilia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.600647. [PMID: 39211207 PMCID: PMC11361019 DOI: 10.1101/2024.07.23.600647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Extracellular vesicles (EVs) mediate intercellular communication during immune responses. EVs are abundant in respiratory biofluids, and the composition of EVs in the lung changes during inflammation. Objective We aimed to quantify the contribution of T cells to airway EVs in allergic lung inflammation and ascertain their function during a type 2 inflammatory response. Methods Genetic membrane tagging was combined with single vesicle flow cytometry to quantify T cell EVs in the airways of mice challenged with ovalbumin or house dust mite. EVs were purified from T helper type 2 (Th2) cell cultures and their functions on eosinophils assessed by flow cytometry and RNA sequencing. Th2 cell EVs were instilled into the lungs of mice to determine effects on lung eosinophilia. Finally, the function of an EV protein cargo was tested using inhibitors and blocking antibodies. Results T cell EVs are increased in the airways of mice with induced allergic inflammation. EVs secreted by Th2 cells inhibit apoptosis and induce activating pathways in eosinophils in vitro. This effect depends on re-stimulation through the T cell receptor. Th2 cell EVs prolong eosinophilia in vivo during allergic airway inflammation. Th2 cell EVs carry a potent form of the cytokine IL-3 on their surfaces, which inhibits apoptosis by activating Jak1/2-dependent pro-survival programs in eosinophils. Conclusion Th2 cell EVs promote eosinophil survival and prolong eosinophilia during allergic airway inflammation. This function depends on the EV cargo IL-3, supporting a role for EVs as vehicles of cytokine-based communication in lung inflammation. Key Messages T cells secrete extracellular vesicles in the airway during allergic lung inflammation.Th2 cell extracellular vesicles inhibit eosinophil apoptosis and prolong airway eosinophilia during allergic lung inflammation.IL-3 carried on Th2 cell EVs is a functional cargo, supporting a role for cytokine-carrying EVs as drivers of type 2 inflammation. Capsule summary This study supports that T cell extracellular vesicles may be important drivers of eosinophilic inflammation through the cytokine cargo IL-3, offering new insights into pro-inflammatory signaling in the allergic lung of patients with asthma.
Collapse
|
167
|
López DA, Griffin A, Aguilar LM, Deering-Rice C, Myers EJ, Warren KJ, Welner RS, Beaudin AE. Prenatal inflammation remodels lung immunity and function by programming ILC2 hyperactivation. Cell Rep 2024; 43:114365. [PMID: 38909363 DOI: 10.1016/j.celrep.2024.114365] [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: 01/11/2024] [Revised: 04/30/2024] [Accepted: 05/31/2024] [Indexed: 06/25/2024] Open
Abstract
Here, we examine how prenatal inflammation shapes tissue function and immunity in the lung by reprogramming tissue-resident immune cells from early development. Maternal, but not fetal, type I interferon-mediated inflammation provokes expansion and hyperactivation of group 2 innate lymphoid cells (ILC2s) seeding the developing lung. Hyperactivated ILC2s produce increased IL-5 and IL-13 and are associated with acute Th2 bias, decreased Tregs, and persistent lung eosinophilia into adulthood. ILC2 hyperactivation is recapitulated by adoptive transfer of fetal liver precursors following prenatal inflammation, indicative of developmental programming at the fetal progenitor level. Reprogrammed ILC2 hyperactivation and subsequent lung immune remodeling, including persistent eosinophilia, is concomitant with worsened histopathology and increased airway dysfunction equivalent to papain exposure, indicating increased asthma susceptibility in offspring. Our data elucidate a mechanism by which early-life inflammation results in increased asthma susceptibility in the presence of hyperactivated ILC2s that drive persistent changes to lung immunity during perinatal development.
Collapse
Affiliation(s)
- Diego A López
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Aleah Griffin
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Lorena Moreno Aguilar
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | | | - Elizabeth J Myers
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | - Kristi J Warren
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Robert S Welner
- Department of Medicine, University of Alabama, Birmingham, AL, USA
| | - Anna E Beaudin
- Department of Pathology, University of Utah, Salt Lake City, UT, USA; Department of Internal Medicine and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
168
|
Garcia-Gonzalez I, Rocha SF, Hamidi A, Garcia-Ortega L, Regano A, Sanchez-Muñoz M, Lytvyn M, Garcia-Cabero A, Roig-Soucase S, Schoofs H, Castro M, Sabata H, Potente M, Graupera M, Makinen T, Benedito R. iSuRe-HadCre is an essential tool for effective conditional genetics. Nucleic Acids Res 2024; 52:e56. [PMID: 38850155 PMCID: PMC11260470 DOI: 10.1093/nar/gkae472] [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: 11/15/2023] [Revised: 05/04/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024] Open
Abstract
Methods for modifying gene function at high spatiotemporal resolution in mice have revolutionized biomedical research, with Cre-loxP being the most widely used technology. However, the Cre-loxP technology has several drawbacks, including weak activity, leakiness, toxicity, and low reliability of existing Cre-reporters. This is mainly because different genes flanked by loxP sites (floxed) vary widely in their sensitivity to Cre-mediated recombination. Here, we report the generation, validation, and utility of iSuRe-HadCre, a new dual Cre-reporter and deleter mouse line that avoids these drawbacks. iSuRe-HadCre achieves this through a novel inducible dual-recombinase genetic cascade that ensures that cells expressing a fluorescent reporter had only transient Cre activity, that is nonetheless sufficient to effectively delete floxed genes. iSuRe-HadCre worked reliably in all cell types and for the 13 floxed genes tested. This new tool will enable the precise, efficient, and trustworthy analysis of gene function in entire mouse tissues or in single cells.
Collapse
Affiliation(s)
- Irene Garcia-Gonzalez
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Susana F Rocha
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Anahita Hamidi
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Lourdes Garcia-Ortega
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alvaro Regano
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Maria S Sanchez-Muñoz
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Mariya Lytvyn
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Aroa Garcia-Cabero
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sergi Roig-Soucase
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Hans Schoofs
- Uppsala University, Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Marco Castro
- Angiogenesis & Metabolism Laboratory, Center of Vascular Biomedicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Helena Sabata
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Michael Potente
- Angiogenesis & Metabolism Laboratory, Center of Vascular Biomedicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mariona Graupera
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
- ICREA, Institució Catalana de Recerca i Estudis Avançats, Pg. Lluís Companys 23, Barcelona, Spain
| | - Taija Makinen
- Uppsala University, Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
- Translational Cancer Medicine Program, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
- Wihuri Research Institute, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Rui Benedito
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
169
|
Sanchez-Vasquez E, Bronner ME, Zernicka-Goetz M. HIF1A contributes to the survival of aneuploid and mosaic pre-implantation embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.04.556218. [PMID: 39071426 PMCID: PMC11275769 DOI: 10.1101/2023.09.04.556218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Human fertility is suboptimal, partly due to error-prone divisions in early cleavage-stages that result in aneuploidy. Most human pre-implantation are mosaics of euploid and aneuploid cells, however, mosaic embryos with a low proportion of aneuploid cells have a similar likelihood of developing to term as fully euploid embryos. How embryos manage aneuploidy during development is poorly understood. This knowledge is crucial for improving fertility treatments and reducing developmental defects. To explore these mechanisms, we established a new mouse model of chromosome mosaicism to study the fate of aneuploid cells during pre-implantation development. We previously used the Mps1 inhibitor reversine to generate aneuploidy in embryos. Here, we found that treatment with the more specific Mps1 inhibitor AZ3146 induced chromosome segregation defects in pre-implantation embryos, similar to reversine. However, AZ3146-treated embryos showed a higher developmental potential than reversine-treated embryos. Unlike reversine-treated embryos, AZ3146-treated embryos exhibited transient upregulation of Hypoxia Inducible-Factor-1A (HIF1A) and lacked p53 upregulation. Pre-implantation embryos develop in a hypoxic environment in vivo, and hypoxia exposure in vitro reduced DNA damage in response to Mps1 inhibition and increased the proportion of euploid cells in the mosaic epiblast. Inhibiting HIF1A in mosaic embryos also decreased the proportion of aneuploid cells in mosaic embryos. Our work illuminates potential strategies to improve the developmental potential of mosaic embryos.
Collapse
Affiliation(s)
| | - Marianne E. Bronner
- Division of Biology 139-74, California Institute of Technology, Pasadena, CA 91125, USA
| | - Magdalena Zernicka-Goetz
- Division of Biology 139-74, California Institute of Technology, Pasadena, CA 91125, USA
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
170
|
Zhu M, Gu B, Thomas EC, Huang Y, Kim YK, Tao H, Yung TM, Chen X, Zhang K, Woolaver EK, Nevin MR, Huang X, Winklbauer R, Rossant J, Sun Y, Hopyan S. A fibronectin gradient remodels mixed-phase mesoderm. SCIENCE ADVANCES 2024; 10:eadl6366. [PMID: 39028807 PMCID: PMC11259159 DOI: 10.1126/sciadv.adl6366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 06/14/2024] [Indexed: 07/21/2024]
Abstract
Physical processes ultimately shape tissue during development. Two emerging proposals are that cells migrate toward stiffer tissue (durotaxis) and that the extent of cell rearrangements reflects tissue phase, but it is unclear whether and how these concepts are related. Here, we identify fibronectin-dependent tissue stiffness as a control variable that underlies and unifies these phenomena in vivo. In murine limb bud mesoderm, cells are either caged, move directionally, or intercalate as a function of their location along a stiffness gradient. A modified Landau phase equation that incorporates tissue stiffness accurately predicts cell diffusivity upon loss or gain of fibronectin. Fibronectin is regulated by WNT5A-YAP feedback that controls cell movements, tissue shape, and skeletal pattern. The results identify a key determinant of phase transition and show how fibronectin-dependent directional cell movement emerges in a mixed-phase environment in vivo.
Collapse
Affiliation(s)
- Min Zhu
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Bin Gu
- Department of Obstetrics Gynecology and Reproductive Biology, and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Evan C. Thomas
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Yunyun Huang
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Yun-Kyo Kim
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Hirotaka Tao
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Theodora M. Yung
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xin Chen
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Kaiwen Zhang
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Elizabeth K. Woolaver
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mikaela R. Nevin
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xi Huang
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Rudolph Winklbauer
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yu Sun
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Orthopaedic Surgery, The Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
171
|
Malinská N, Grobárová V, Knížková K, Černý J. Maternal-Fetal Microchimerism: Impacts on Offspring's Immune Development and Transgenerational Immune Memory Transfer. Physiol Res 2024; 73:315-332. [PMID: 39027950 PMCID: PMC11299782 DOI: 10.33549/physiolres.935296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/06/2024] [Indexed: 07/27/2024] Open
Abstract
Maternal-fetal microchimerism is a fascinating phenomenon in which maternal cells migrate to the tissues of the offspring during both pregnancy and breastfeeding. These cells primarily consist of leukocytes and stem cells. Remarkably, these maternal cells possess functional potential in the offspring and play a significant role in shaping their immune system development. T lymphocytes, a cell population mainly found in various tissues of the offspring, have been identified as the major cell type derived from maternal microchimerism. These T lymphocytes not only exert effector functions but also influence the development of the offspring's T lymphocytes in the thymus and the maturation of B lymphocytes in the lymph nodes. Furthermore, the migration of maternal leukocytes also facilitates the transfer of immune memory across generations. Maternal microchimerism has also been observed to address immunodeficiencies in the offspring. This review article focuses on investigating the impact of maternal cells transported within maternal microchimerism on the immune system development of the offspring, as well as elucidating the effector functions of maternal cells that migrate through the placenta and breast milk to reach the offspring.
Collapse
Affiliation(s)
- N Malinská
- Laboratory of Cell Immunology, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic.
| | | | | | | |
Collapse
|
172
|
Home P, Ghosh A, Kumar RP, Ray S, Gunewardena S, Kumar R, Dasgupta P, Roy N, Saha A, Ouseph MM, Leone GW, Paul S. A Single Trophoblast Layer Acts as the Gatekeeper at the Endothelial-Hematopoietic Crossroad in the Placenta. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603303. [PMID: 39071312 PMCID: PMC11275844 DOI: 10.1101/2024.07.12.603303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
During embryonic development the placental vasculature acts as a major hematopoietic niche, where endothelial to hematopoietic transition ensures emergence of hematopoietic stem cells (HSCs). However, the molecular mechanisms that regulate the placental hematoendothelial niche are poorly understood. Using a parietal trophoblast giant cell (TGC)-specific knockout mouse model and single-cell RNA-sequencing, we show that the paracrine factors secreted by the TGCs are critical in the development of this niche. Disruptions in the TGC-specific paracrine signaling leads to the loss of HSC population and the concomitant expansion of a KDR+/DLL4+/PROM1+ hematoendothelial cell-population in the placenta. Combining single-cell transcriptomics and receptor-ligand pair analyses, we also define the parietal TGC-dependent paracrine signaling network and identify Integrin signaling as a fundamental regulator of this process. Our study elucidates novel mechanisms by which non-autonomous signaling from the primary parietal TGCs maintain the delicate placental hematopoietic-angiogenic balance and ensures embryonic and extraembryonic development.
Collapse
Affiliation(s)
- Pratik Home
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Present address: XenoTech, A BioIVT Company, 1101 W Cambridge Cir Dr, Kansas City, KS 66103
| | - Ananya Ghosh
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Present address: Department of Urology, University of California San Francisco, 35, Medical 12 Center Way, San Francisco, CA 94143
| | - Ram Parikshan Kumar
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Institute for Reproductive Health and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Soma Ray
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Rajnish Kumar
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Purbasa Dasgupta
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Namrata Roy
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Abhik Saha
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Madhu M. Ouseph
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065
| | - Gustavo W. Leone
- Department of Biochemistry, Medical College of Wisconsin, WI 53226, USA
| | - Soumen Paul
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Institute for Reproductive Health and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
173
|
Guo M, Wu Y, Hobson CM, Su Y, Qian S, Krueger E, Christensen R, Kroeschell G, Bui J, Chaw M, Zhang L, Liu J, Hou X, Han X, Lu Z, Ma X, Zhovmer A, Combs C, Moyle M, Yemini E, Liu H, Liu Z, Benedetto A, La Riviere P, Colón-Ramos D, Shroff H. Deep learning-based aberration compensation improves contrast and resolution in fluorescence microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.15.562439. [PMID: 37986950 PMCID: PMC10659418 DOI: 10.1101/2023.10.15.562439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Optical aberrations hinder fluorescence microscopy of thick samples, reducing image signal, contrast, and resolution. Here we introduce a deep learning-based strategy for aberration compensation, improving image quality without slowing image acquisition, applying additional dose, or introducing more optics into the imaging path. Our method (i) introduces synthetic aberrations to images acquired on the shallow side of image stacks, making them resemble those acquired deeper into the volume and (ii) trains neural networks to reverse the effect of these aberrations. We use simulations and experiments to show that applying the trained 'de-aberration' networks outperforms alternative methods, providing restoration on par with adaptive optics techniques; and subsequently apply the networks to diverse datasets captured with confocal, light-sheet, multi-photon, and super-resolution microscopy. In all cases, the improved quality of the restored data facilitates qualitative image inspection and improves downstream image quantitation, including orientational analysis of blood vessels in mouse tissue and improved membrane and nuclear segmentation in C. elegans embryos.
Collapse
|
174
|
Gillespie W, Zhang Y, Ruiz OE, Cerda J, Ortiz-Guzman J, Turner WD, Largoza G, Sherman M, Mosser LE, Fujimoto E, Chien CB, Kwan KM, Arenkiel BR, Devine WP, Wythe JD. Multisite Assembly of Gateway Induced Clones (MAGIC): a flexible cloning toolbox with diverse applications in vertebrate model systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.13.603267. [PMID: 39026881 PMCID: PMC11257631 DOI: 10.1101/2024.07.13.603267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Here we present the Multisite Assembly of Gateway Induced Clones (MAGIC) system, which harnesses site-specific recombination-based cloning via Gateway technology for rapid, modular assembly of between 1 and 3 "Entry" vector components, all into a fourth, standard high copy "Destination" plasmid backbone. The MAGIC toolkit spans a range of in vitro and in vivo uses, from directing tunable gene expression, to driving simultaneous expression of microRNAs and fluorescent reporters, to enabling site-specific recombinase-dependent gene expression. All MAGIC system components are directly compatible with existing multisite gateway Tol2 systems currently used in zebrafish, as well as existing eukaryotic cell culture expression Destination plasmids, and available mammalian lentiviral and adenoviral Destination vectors, allowing rapid cross-species experimentation. Moreover, herein we describe novel vectors with flanking piggyBac transposon elements for stable genomic integration in vitro or in vivo when used with piggyBac transposase. Collectively, the MAGIC system facilitates transgenesis in cultured mammalian cells, electroporated mouse and chick embryos, as well as in injected zebrafish embryos, enabling the rapid generation of innovative DNA constructs for biological research due to a shared, common plasmid platform.
Collapse
|
175
|
Wang Y, Heymann F, Peiseler M. Intravital imaging: dynamic insights into liver immunity in health and disease. Gut 2024; 73:1364-1375. [PMID: 38777574 DOI: 10.1136/gutjnl-2023-331739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
Inflammation is a critical component of most acute and chronic liver diseases. The liver is a unique immunological organ with a dense vascular network, leading to intense crosstalk between tissue-resident immune cells, passenger leucocytes and parenchymal cells. During acute and chronic liver diseases, the multifaceted immune response is involved in disease promoting and repair mechanisms, while upholding core liver immune functions. In recent years, single-cell technologies have unravelled a previously unknown heterogeneity of immune cells, reshaping the complexity of the hepatic immune response. However, inflammation is a dynamic biological process, encompassing various immune cells, orchestrated in temporal and spatial dimensions, and driven by multiorgan signals. Intravital microscopy (IVM) has emerged as a powerful tool to investigate immunity by visualising the dynamic interplay between different immune cells and their surroundings within a near-natural environment. In this review, we summarise the experimental considerations to perform IVM and highlight recent technological developments. Furthermore, we outline the unique contributions of IVM to our understanding of liver immunity. Through the lens of liver disease, we discuss novel immune-mediated disease mechanisms uncovered by imaging-based studies.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Heymann
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz Peiseler
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité, Berlin, Germany
| |
Collapse
|
176
|
LeBourdais R, Grifno GN, Banerji R, Regan K, Suki B, Nia HT. Mapping the strain-stiffening behavior of the lung and lung cancer at microscale resolution using the crystal ribcage. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1396593. [PMID: 39050550 PMCID: PMC11266057 DOI: 10.3389/fnetp.2024.1396593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/10/2024] [Indexed: 07/27/2024]
Abstract
Lung diseases such as cancer substantially alter the mechanical properties of the organ with direct impact on the development, progression, diagnosis, and treatment response of diseases. Despite significant interest in the lung's material properties, measuring the stiffness of intact lungs at sub-alveolar resolution has not been possible. Recently, we developed the crystal ribcage to image functioning lungs at optical resolution while controlling physiological parameters such as air pressure. Here, we introduce a data-driven, multiscale network model that takes images of the lung at different distending pressures, acquired via the crystal ribcage, and produces corresponding absolute stiffness maps. Following validation, we report absolute stiffness maps of the functioning lung at microscale resolution in health and disease. For representative images of a healthy lung and a lung with primary cancer, we find that while the lung exhibits significant stiffness heterogeneity at the microscale, primary tumors introduce even greater heterogeneity into the lung's microenvironment. Additionally, we observe that while the healthy alveoli exhibit strain-stiffening of ∼1.75 times, the tumor's stiffness increases by a factor of six across the range of measured transpulmonary pressures. While the tumor stiffness is 1.4 times the lung stiffness at a transpulmonary pressure of three cmH2O, the tumor's mean stiffness is nearly five times greater than that of the surrounding tissue at a transpulmonary pressure of 18 cmH2O. Finally, we report that the variance in both strain and stiffness increases with transpulmonary pressure in both the healthy and cancerous lungs. Our new method allows quantitative assessment of disease-induced stiffness changes in the alveoli with implications for mechanotransduction.
Collapse
Affiliation(s)
| | | | | | | | | | - Hadi T. Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
177
|
Ren SY, Xia Y, Yu B, Lei QJ, Hou PF, Guo S, Wu SL, Liu W, Yang SF, Jiang YB, Chen JF, Shen KF, Zhang CQ, Wang F, Yan M, Ren H, Yang N, Zhang J, Zhang K, Lin S, Li T, Yang QW, Xiao L, Hu ZX, Mei F. Growth hormone promotes myelin repair after chronic hypoxia via triggering pericyte-dependent angiogenesis. Neuron 2024; 112:2177-2196.e6. [PMID: 38653248 DOI: 10.1016/j.neuron.2024.03.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/26/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
White matter injury (WMI) causes oligodendrocyte precursor cell (OPC) differentiation arrest and functional deficits, with no effective therapies to date. Here, we report increased expression of growth hormone (GH) in the hypoxic neonatal mouse brain, a model of WMI. GH treatment during or post hypoxic exposure rescues hypoxia-induced hypomyelination and promotes functional recovery in adolescent mice. Single-cell sequencing reveals that Ghr mRNA expression is highly enriched in vascular cells. Cell-lineage labeling and tracing identify the GHR-expressing vascular cells as a subpopulation of pericytes. These cells display tip-cell-like morphology with kinetic polarized filopodia revealed by two-photon live imaging and seemingly direct blood vessel branching and bridging. Gain-of-function and loss-of-function experiments indicate that GHR signaling in pericytes is sufficient to modulate angiogenesis in neonatal brains, which enhances OPC differentiation and myelination indirectly. These findings demonstrate that targeting GHR and/or downstream effectors may represent a promising therapeutic strategy for WMI.
Collapse
Affiliation(s)
- Shu-Yu Ren
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu Xia
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Yu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qi-Jing Lei
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Peng-Fei Hou
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Sheng Guo
- Department of Immunology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shuang-Ling Wu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wei Liu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shao-Fan Yang
- Brain Research Center, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yi-Bin Jiang
- Department of Neurobiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing-Fei Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kai-Feng Shen
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chun-Qing Zhang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Fei Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mi Yan
- Department of Pediatrics, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400000, China
| | - Hong Ren
- Department of Emergence, 5(th) People's Hospital of Chongqing, Chongqing 400062, China
| | - Nian Yang
- Department of Physiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Zhang
- Department of Neurobiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kuan Zhang
- Brain Research Center, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Sen Lin
- Department of Neurology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tao Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhang-Xue Hu
- Department of Pediatrics, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400000, China.
| | - Feng Mei
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
178
|
Qian Q, Li M, Zhang Z, Davis SW, Rahmouni K, Norris AW, Cao H, Ding WX, Hotamisligil GS, Yang L. Obesity disrupts the pituitary-hepatic UPR communication leading to NAFLD progression. Cell Metab 2024; 36:1550-1565.e9. [PMID: 38718793 PMCID: PMC11222033 DOI: 10.1016/j.cmet.2024.04.014] [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: 08/17/2023] [Revised: 03/05/2024] [Accepted: 04/17/2024] [Indexed: 07/05/2024]
Abstract
Obesity alters levels of pituitary hormones that govern hepatic immune-metabolic homeostasis, dysregulation of which leads to nonalcoholic fatty liver disease (NAFLD). However, the impact of obesity on intra-pituitary homeostasis is largely unknown. Here, we uncovered a blunted unfolded protein response (UPR) but elevated inflammatory signatures in pituitary glands of obese mice and humans. Furthermore, we found that obesity inflames the pituitary gland, leading to impaired pituitary inositol-requiring enzyme 1α (IRE1α)-X-box-binding protein 1 (XBP1) UPR branch, which is essential for protecting against pituitary endocrine defects and NAFLD progression. Intriguingly, pituitary IRE1-deletion resulted in hypothyroidism and suppressed the thyroid hormone receptor B (THRB)-mediated activation of Xbp1 in the liver. Conversely, activation of the hepatic THRB-XBP1 axis improved NAFLD in mice with pituitary UPR defect. Our study provides the first evidence and mechanism of obesity-induced intra-pituitary cellular defects and the pathophysiological role of pituitary-liver UPR communication in NAFLD progression.
Collapse
Affiliation(s)
- Qingwen Qian
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Mark Li
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zeyuan Zhang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Shannon W Davis
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Andrew W Norris
- Division of Endocrinology and Diabetes, Department of Pediatrics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Huojun Cao
- Iowa Institute for Oral Health Research, Division of Biostatistics and Computational Biology, Department of Endodontics, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. School of Public Health, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02141, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
179
|
Shao F, Phan AV, Yu W, Guo Y, Thompson J, Coppinger C, Venugopalan SR, Amendt BA, Van Otterloo E, Cao H. Transcriptional programs of Pitx2 and Tfap2a/Tfap2b controlling lineage specification of mandibular epithelium during tooth initiation. PLoS Genet 2024; 20:e1011364. [PMID: 39052671 PMCID: PMC11302917 DOI: 10.1371/journal.pgen.1011364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 08/06/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
How the dorsal-ventral axis of the vertebrate jaw, particularly the position of tooth initiation site, is established remains a critical and unresolved question. Tooth development starts with the formation of the dental lamina, a localized thickened strip within the maxillary and mandibular epithelium. To identify transcriptional regulatory networks (TRN) controlling the specification of dental lamina from the naïve mandibular epithelium, we utilized Laser Microdissection coupled low-input RNA-seq (LMD-RNA-seq) to profile gene expression of different domains of the mandibular epithelium along the dorsal-ventral axis. We comprehensively identified transcription factors (TFs) and signaling pathways that are differentially expressed along mandibular epithelial domains (including the dental lamina). Specifically, we found that the TFs Sox2 and Tfap2 (Tfap2a/Tfap2b) formed complimentary expression domains along the dorsal-ventral axis of the mandibular epithelium. Interestingly, both classic and novel dental lamina specific TFs-such as Pitx2, Ascl5 and Zfp536-were found to localize near the Sox2:Tfap2a/Tfap2b interface. To explore the functional significance of these domain specific TFs, we next examined loss-of-function mouse models of these domain specific TFs, including the dental lamina specific TF, Pitx2, and the ventral surface ectoderm specific TFs Tfap2a and Tfap2b. We found that disruption of domain specific TFs leads to an upregulation and expansion of the alternative domain's TRN. The importance of this cross-repression is evident by the ectopic expansion of Pitx2 and Sox2 positive dental lamina structure in Tfap2a/Tfap2b ectodermal double knockouts and the emergence of an ectopic tooth in the ventral surface ectoderm. Finally, we uncovered an unappreciated interface of mesenchymal SHH and WNT signaling pathways, at the site of tooth initiation, that were established by the epithelial domain specific TFs including Pitx2 and Tfap2a/Tfap2b. These results uncover a previously unknown molecular mechanism involving cross-repression of domain specific TFs including Pitx2 and Tfap2a/Tfap2b in patterning the dorsal-ventral axis of the mouse mandible, specifically the regulation of tooth initiation site.
Collapse
Affiliation(s)
- Fan Shao
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - An-Vi Phan
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
| | - Wenjie Yu
- Department of Internal Medicine and Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Yuwei Guo
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
| | - Jamie Thompson
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Carter Coppinger
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
| | - Shankar R. Venugopalan
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
- Department of Orthodontics, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
| | - Brad A. Amendt
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Department of Orthodontics, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
| | - Eric Van Otterloo
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Department of Periodontics, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
| | - Huojun Cao
- Iowa Institute for Oral Health Research, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Division of Biostatistics and Computational Biology, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
- Department of Endodontics, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, United States of America
| |
Collapse
|
180
|
Mark Kim MK, Lawrence M, Quinonez D, Brooks C, Ramachandran R, Séguin CA. Transient receptor potential vanilloid 4 regulates extracellular matrix composition and mediates load-induced intervertebral disc degeneration in a mouse model. Osteoarthritis Cartilage 2024; 32:881-894. [PMID: 38604493 DOI: 10.1016/j.joca.2024.04.001] [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: 10/18/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/13/2024]
Abstract
OBJECTIVE Transient receptor potential vanilloid 4 (TRPV4) is a multi-modally activated cation channel that mediates mechanotransduction pathways by which musculoskeletal tissues respond to mechanical load and regulate tissue health. Using conditional Trpv4 knockout mice, we investigated the role of Trpv4 in regulating intervertebral disc (IVD) health and injury-induced IVD degeneration. METHODS Col2-Cre;Trpv4fl/f (Trpv4 KO) mice were used to knockout Trpv4 in all type 2 collagen-expressing cells. Effects of gene targeting alone was assessed in lumbar spines, using vertebral bone length measurement, histological, immunohistochemistry and gene expression analyses, and mechanical testing. Disc puncture was performed on caudal IVDs of wild-type (WT) and Trpv4 KO mice at 2.5- and 6.5-months-of-age. Six weeks after puncture (4- and 8-months-of-age at sacrifice), caudal spines were assessed using histological analyses. RESULTS While loss of Trpv4 did not significantly alter vertebral bone length and tissue histomorphology compared to age-matched WT mice, Trpv4 KO mice showed decreased proteoglycan and PRG4 staining in the annulus fibrosus compared to WT. At the gene level, Trpv4 KO mice showed significantly increased expression of Acan, Bgn, and Prg4 compared to WT. Functionally, loss of Trpv4 was associated with significantly increased neutral zone length in lumbar IVDs. Following puncture, both Trpv4 KO and WT mice showed similar signs of degeneration at the site of injury. Interestingly, loss of Trpv4 prevented mechanically-induced degeneration in IVDs adjacent to sites of injury. CONCLUSION These studies suggest a role for Trpv4 in regulating extracellular matrix synthesis and mediating the response of IVD tissues to mechanical stress.
Collapse
Affiliation(s)
- Min Kyu Mark Kim
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Matthew Lawrence
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Diana Quinonez
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Courtney Brooks
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Cheryle A Séguin
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
181
|
Ma R, Feng D, Chen J, Zhou J, Xia K, Kong X, Hu G, Lu P. Targeting Tumor Heterogeneity by Breaking a Stem Cell and Epithelial Niche Interaction Loop. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307452. [PMID: 38708713 PMCID: PMC11234407 DOI: 10.1002/advs.202307452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/20/2024] [Indexed: 05/07/2024]
Abstract
Tumor heterogeneity, the presence of multiple distinct subpopulations of cancer cells between patients or among the same tumors, poses a major challenge to current targeted therapies. The way these different subpopulations interact among themselves and the stromal niche environment, and how such interactions affect cancer stem cell behavior has remained largely unknown. Here, it is shown that an FGF-BMP7-INHBA signaling positive feedback loop integrates interactions among different cell populations, including mammary gland stem cells, luminal epithelial and stromal fibroblast niche components not only in organ regeneration but also, with certain modifications, in cancer progression. The reciprocal dependence of basal stem cells and luminal epithelium is based on basal-derived BMP7 and luminal-derived INHBA, which promote their respective expansion, and is regulated by stromal-epithelial FGF signaling. Targeting this interaction loop, for example, by reducing the function of one or more of its components, inhibits organ regeneration and breast cancer progression. The results have profound implications for overcoming drug resistance because of tumor heterogeneity in future targeted therapies.
Collapse
Affiliation(s)
- Rongze Ma
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Hengyang, Hunan, 421001, China
- Institute of Cell Biology, University of South China, Hengyang, Hunan, 421001, China
- Institute for Future Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Deyi Feng
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Hengyang, Hunan, 421001, China
- Institute of Cell Biology, University of South China, Hengyang, Hunan, 421001, China
| | - Jing Chen
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201210, China
| | - Jiecan Zhou
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Kun Xia
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Hengyang, Hunan, 421001, China
- Institute of Cell Biology, University of South China, Hengyang, Hunan, 421001, China
| | - Xiangyin Kong
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Guohong Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Pengfei Lu
- MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Hengyang, Hunan, 421001, China
- Institute of Cell Biology, University of South China, Hengyang, Hunan, 421001, China
- Institute for Future Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
182
|
Xu N, Alfieri CM, Yu Y, Guo M, Yutzey KE. Wnt Signaling Inhibition Prevents Postnatal Inflammation and Disease Progression in Mouse Congenital Myxomatous Valve Disease. Arterioscler Thromb Vasc Biol 2024; 44:1540-1554. [PMID: 38660802 PMCID: PMC11209782 DOI: 10.1161/atvbaha.123.320388] [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: 11/06/2023] [Accepted: 04/10/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Myxomatous valve disease (MVD) is the most common cause of mitral regurgitation, leading to impaired cardiac function and heart failure. MVD in a mouse model of Marfan syndrome includes valve leaflet thickening and progressive valve degeneration. However, the underlying mechanisms by which the disease progresses remain undefined. METHODS Mice with Fibrillin 1 gene variant Fbn1C1039G/+ recapitulate histopathologic features of Marfan syndrome, and Wnt (Wingless-related integration site) signaling activity was detected in TCF/Lef-lacZ (T-cell factor/lymphoid enhancer factor-β-galactosidase) reporter mice. Single-cell RNA sequencing was performed from mitral valves of wild-type and Fbn1C1039G/+ mice at 1 month of age. Inhibition of Wnt signaling was achieved by conditional induction of the secreted Wnt inhibitor Dkk1 (Dickkopf-1) expression in periostin-expressing valve interstitial cells of Periostin-Cre; tetO-Dkk1; R26rtTA; TCF/Lef-lacZ; Fbn1C1039G/+ mice. Dietary doxycycline was administered for 1 month beginning with MVD initiation (1-month-old) or MVD progression (2-month-old). Histological evaluation and immunofluorescence for ECM (extracellular matrix) and immune cells were performed. RESULTS Wnt signaling is activated early in mitral valve disease progression, before immune cell infiltration in Fbn1C1039G/+ mice. Single-cell transcriptomics revealed similar mitral valve cell heterogeneity between wild-type and Fbn1C1039G/+ mice at 1 month of age. Wnt pathway genes were predominantly expressed in valve interstitial cells and valve endothelial cells of Fbn1C1039G/+ mice. Inhibition of Wnt signaling in Fbn1C1039G/+ mice at 1 month of age prevented the initiation of MVD as indicated by improved ECM remodeling and reduced valve leaflet thickness with decreased infiltrating macrophages. However, later, Wnt inhibition starting at 2 months did not prevent the progression of MVD. CONCLUSIONS Wnt signaling is involved in the initiation of mitral valve abnormalities and inflammation but is not responsible for later-stage valve disease progression once it has been initiated. Thus, Wnt signaling contributes to MVD progression in a time-dependent manner and provides a promising therapeutic target for the early treatment of congenital MVD in Marfan syndrome.
Collapse
Affiliation(s)
- Na Xu
- Division of Molecular Cardiovascular Biology, the Heart Institute, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Christina M. Alfieri
- Division of Molecular Cardiovascular Biology, the Heart Institute, Cincinnati Children’s Hospital Medical Center
| | - Yang Yu
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center
| | - Minzhe Guo
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Katherine E. Yutzey
- Division of Molecular Cardiovascular Biology, the Heart Institute, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| |
Collapse
|
183
|
Zhuang X, Wang Q, Joost S, Ferrena A, Humphreys DT, Li Z, Blum M, Bastl K, Ding S, Landais Y, Zhan Y, Zhao Y, Chaligne R, Lee JH, Carrasco SE, Bhanot UK, Koche RP, Bott MJ, Katajisto P, Soto-Feliciano YM, Pisanic T, Thomas T, Zheng D, Wong ES, Tammela T. Aging limits stemness and tumorigenesis in the lung by reprogramming iron homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.23.600305. [PMID: 38979280 PMCID: PMC11230188 DOI: 10.1101/2024.06.23.600305] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Aging is associated with a decline in the number and fitness of adult stem cells 1-4 . Aging-associated loss of stemness is posited to suppress tumorigenesis 5,6 , but this hypothesis has not been tested in vivo . Here, using physiologically aged autochthonous genetically engineered mouse models and primary cells 7,8 , we demonstrate aging suppresses lung cancer initiation and progression by degrading stemness of the alveolar cell of origin. This phenotype is underpinned by aging-associated induction of the transcription factor NUPR1 and its downstream target lipocalin-2 in the cell of origin in mice and humans, leading to a functional iron insufficiency in the aged cells. Genetic inactivation of the NUPR1-lipocalin-2 axis or iron supplementation rescue stemness and promote tumorigenic potential of aged alveolar cells. Conversely, targeting the NUPR1- lipocalin-2 axis is detrimental to young alveolar cells via induction of ferroptosis. We find that aging-associated DNA hypomethylation at specific enhancer sites associates with elevated NUPR1 expression, which is recapitulated in young alveolar cells by inhibition of DNA methylation. We uncover that aging drives a functional iron insufficiency, which leads to loss of stemness and tumorigenesis, but promotes resistance to ferroptosis. These findings have significant implications for the therapeutic modulation of cellular iron homeostasis in regenerative medicine and in cancer prevention. Furthermore, our findings are consistent with a model whereby most human cancers initiate in young individuals, revealing a critical window for such cancer prevention efforts.
Collapse
|
184
|
Perrin S, Protic S, Bretegnier V, Laurendeau I, de Lageneste OD, Panara N, Ruckebusch O, Luka M, Masson C, Maillard T, Coulpier F, Pannier S, Wicart P, Hadj-Rabia S, Radomska KJ, Zarhrate M, Ménager M, Vidaud D, Topilko P, Parfait B, Colnot C. MEK-SHP2 inhibition prevents tibial pseudarthrosis caused by NF1 loss in Schwann cells and skeletal stem/progenitor cells. Sci Transl Med 2024; 16:eadj1597. [PMID: 38924432 DOI: 10.1126/scitranslmed.adj1597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/15/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
Congenital pseudarthrosis of the tibia (CPT) is a severe pathology marked by spontaneous bone fractures that fail to heal, leading to fibrous nonunion. Half of patients with CPT are affected by the multisystemic genetic disorder neurofibromatosis type 1 (NF1) caused by mutations in the NF1 tumor suppressor gene, a negative regulator of RAS-mitogen-activated protein kinase (MAPK) signaling pathway. Here, we analyzed patients with CPT and Prss56-Nf1 knockout mice to elucidate the pathogenic mechanisms of CPT-related fibrous nonunion and explored a pharmacological approach to treat CPT. We identified NF1-deficient Schwann cells and skeletal stem/progenitor cells (SSPCs) in pathological periosteum as affected cell types driving fibrosis. Whereas NF1-deficient SSPCs adopted a fibrotic fate, NF1-deficient Schwann cells produced critical paracrine factors including transforming growth factor-β and induced fibrotic differentiation of wild-type SSPCs. To counteract the elevated RAS-MAPK signaling in both NF1-deficient Schwann cells and SSPCs, we used MAPK kinase (MEK) and Src homology 2 containing protein tyrosine phosphatase 2 (SHP2) inhibitors. Combined MEK-SHP2 inhibition in vivo prevented fibrous nonunion in the Prss56-Nf1 knockout mouse model, providing a promising therapeutic strategy for the treatment of fibrous nonunion in CPT.
Collapse
Affiliation(s)
- Simon Perrin
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Sanela Protic
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | | | - Ingrid Laurendeau
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
| | | | - Nicolas Panara
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
| | - Odile Ruckebusch
- Université Paris Est Creteil, INSERM, IMRB, Plateforme de Cytométrie en flux, 94000 Creteil, France
| | - Marine Luka
- Paris Cité University, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Cécile Masson
- Bioinformatics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163, 75015 Paris, France
- INSERM US24/CNRS UAR3633, Paris Cité University, 75015 Paris, France
| | - Théodora Maillard
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Fanny Coulpier
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Stéphanie Pannier
- Department of Pediatric Orthopedic Surgery and Traumatology, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | - Philippe Wicart
- Department of Pediatric Orthopedic Surgery and Traumatology, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | - Smail Hadj-Rabia
- Department of Dermatology, Reference Center for Rare Skin Diseases (MAGEC), Imagine Institute, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | | | - Mohammed Zarhrate
- INSERM US24/CNRS UAR3633, Paris Cité University, 75015 Paris, France
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163, 75015 Paris, France
| | - Mickael Ménager
- Paris Cité University, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Dominique Vidaud
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Piotr Topilko
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Béatrice Parfait
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Céline Colnot
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| |
Collapse
|
185
|
Jimenez-Cyrus D, Adusumilli VS, Stempel MH, Maday S, Ming GL, Song H, Bond AM. Molecular cascade reveals sequential milestones underlying hippocampal neural stem cell development into an adult state. Cell Rep 2024; 43:114339. [PMID: 38852158 PMCID: PMC11320877 DOI: 10.1016/j.celrep.2024.114339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 04/16/2024] [Accepted: 05/23/2024] [Indexed: 06/11/2024] Open
Abstract
Quiescent adult neural stem cells (NSCs) in the mammalian brain arise from proliferating NSCs during development. Beyond acquisition of quiescence, an adult NSC hallmark, little is known about the process, milestones, and mechanisms underlying the transition of developmental NSCs to an adult NSC state. Here, we performed targeted single-cell RNA-seq analysis to reveal the molecular cascade underlying NSC development in the early postnatal mouse dentate gyrus. We identified two sequential steps, first a transition to quiescence followed by further maturation, each of which involved distinct changes in metabolic gene expression. Direct metabolic analysis uncovered distinct milestones, including an autophagy burst before NSC quiescence acquisition and cellular reactive oxygen species level elevation along NSC maturation. Functionally, autophagy is important for the NSC transition to quiescence during early postnatal development. Together, our study reveals a multi-step process with defined milestones underlying establishment of the adult NSC pool in the mammalian brain.
Collapse
Affiliation(s)
- Dennisse Jimenez-Cyrus
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vijay S Adusumilli
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Max H Stempel
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandra Maday
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; The Epigenetics Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Allison M Bond
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
186
|
Yang N, Sun Y, Han B, Deng N, Li G, Han Q, Wang Y, Cai H, Liu F, Cao B, Deng W, Bao H, Kong S, Lu J, Wang H. Trophoblastic signals facilitate endometrial interferon response and lipid metabolism, ensuring normal decidualization. Cell Rep 2024; 43:114246. [PMID: 38762885 DOI: 10.1016/j.celrep.2024.114246] [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: 11/03/2023] [Revised: 04/04/2024] [Accepted: 05/02/2024] [Indexed: 05/21/2024] Open
Abstract
The decidua plays a crucial role in providing structural and trophic support to the developing conceptus before placentation. Following embryo attachment, embryonic components intimately interact with the decidual tissue. While evidence indicates the participation of embryo-derived factors in crosstalk with the uterus, the extent of their impact on post-implantation decidual development requires further investigation. Here, we utilize transgenic mouse models to selectively eliminate primary trophoblast giant cells (pTGCs), the embryonic cells that interface with maternal tissue at the forefront. pTGC ablation impairs decidualization and compromises decidual interferon response and lipid metabolism. Mechanistically, pTGCs release factors such as interferon kappa (IFNK) to strengthen the decidual interferon response and lipoprotein lipase (LPL) to enhance lipid accumulation within the decidua, thereby promoting decidualization. This study presents genetic and metabolomic evidence reinforcing the proactive role of pTGC-derived factors in mobilizing maternal resources to strengthen decidualization, facilitating the normal progression of early pregnancy.
Collapse
Affiliation(s)
- Ningjie Yang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yang Sun
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Bing Han
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Na Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Gaizhen Li
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Qian Han
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yinan Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Han Cai
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Fan Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Bin Cao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Wenbo Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Haili Bao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
187
|
Yang M, Shulkin N, Gonzalez E, Castillo J, Yan C, Zhang K, Arvanitis L, Borok Z, Wallace WD, Raz D, Torres ETR, Marconett CN. Cell of origin alters myeloid-mediated immunosuppression in lung adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.19.599651. [PMID: 38948812 PMCID: PMC11213232 DOI: 10.1101/2024.06.19.599651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Solid carcinomas are often highly heterogenous cancers, arising from multiple epithelial cells of origin. Yet, how the cell of origin influences the response of the tumor microenvironment is poorly understood. Lung adenocarcinoma (LUAD) arises in the distal alveolar epithelium which is populated primarily by alveolar epithelial type I (AT1) and type II (AT2) cells. It has been previously reported that Gramd2 + AT1 cells can give rise to a histologically-defined LUAD that is distinct in pathology and transcriptomic identity from that arising from Sftpc + AT2 cells1,2. To determine how cells of origin influence the tumor immune microenvironment (TIME) landscape, we comprehensively characterized transcriptomic, molecular, and cellular states within the TIME of Gramd2 + AT1 and Sftpc + AT2-derived LUAD using KRASG12D oncogenic driver mouse models. Myeloid cells within the Gramd2 + AT1-derived LUAD TIME were increased, specifically, immunoreactive monocytes and tumor associated macrophages (TAMs). In contrast, the Sftpc + AT2 LUAD TIME was enriched for Arginase-1+ myeloid derived suppressor cells (MDSC) and TAMs expressing profiles suggestive of immunosuppressive function. Validation of immune infiltration was performed using flow cytometry, and intercellular interaction analysis between the cells of origin and major myeloid cell populations indicated that cell-type specific markers SFTPD in AT2 cells and CAV1 in AT1 cells mediated unique interactions with myeloid cells of the differential immunosuppressive states within each cell of origin mouse model. Taken together, Gramd2 + AT1-derived LUAD presents with an anti-tumor, immunoreactive TIME, while the TIME of Sftpc + AT2-derived LUAD has hallmarks of immunosuppression. This study suggests that LUAD cell of origin influences the composition and suppression status of the TIME landscape and may hold critical implications for patient response to immunotherapy.
Collapse
Affiliation(s)
- Minxiao Yang
- Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA USA 91010
- Department of Surgery, University of Southern California, Los Angeles, CA USA 90089
- Department of Translational Genomics, University of Southern California, Los Angeles, CA USA 90089
| | - Noah Shulkin
- Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA USA 91010
| | - Edgar Gonzalez
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA USA 90089
| | - Jonathan Castillo
- Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA USA 91010
| | - Chunli Yan
- Department of Surgery, University of Southern California, Los Angeles, CA USA 90089
| | - Keqiang Zhang
- Division of Thoracic Surgery, Department of Surgery, City of Hope National Medical Center, City of Hope, Duarte, CA USA 91010
| | - Leonidas Arvanitis
- Department of Pathology, City of Hope National Medical Center, City of Hope, Duarte, CA USA 91010
| | - Zea Borok
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA USA 92093
| | - W. Dean Wallace
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA USA 90089
| | - Dan Raz
- Division of Thoracic Surgery, Department of Surgery, City of Hope National Medical Center, City of Hope, Duarte, CA USA 91010
| | - Evanthia T. Roussos Torres
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA USA 90089
| | - Crystal N. Marconett
- Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA USA 91010
- Department of Surgery, University of Southern California, Los Angeles, CA USA 90089
- Department of Translational Genomics, University of Southern California, Los Angeles, CA USA 90089
| |
Collapse
|
188
|
Zutshi N, Mohapatra BC, Mondal P, An W, Goetz BT, Wang S, Li S, Storck MD, Mercer DF, Black AR, Thayer SP, Black JD, Lin C, Band V, Band H. Cbl and Cbl-b ubiquitin ligases are essential for intestinal epithelial stem cell maintenance. iScience 2024; 27:109912. [PMID: 38974465 PMCID: PMC11225835 DOI: 10.1016/j.isci.2024.109912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 02/29/2024] [Accepted: 05/03/2024] [Indexed: 07/09/2024] Open
Abstract
Receptor tyrosine kinases (RTKs) control stem cell maintenance vs. differentiation decisions. Casitas B-lineage lymphoma (CBL) family ubiquitin ligases are negative regulators of RTKs, but their stem cell regulatory roles remain unclear. Here, we show that Lgr5+ intestinal stem cell (ISC)-specific inducible Cbl-knockout (KO) on a Cblb null mouse background (iDKO) induced rapid loss of the Lgr5 Hi ISCs with transient expansion of the Lgr5 Lo transit-amplifying population. LacZ-based lineage tracing revealed increased ISC commitment toward enterocyte and goblet cell fate at the expense of Paneth cells. Functionally, Cbl/Cblb iDKO impaired the recovery from radiation-induced intestinal epithelial injury. In vitro, Cbl/Cblb iDKO led to inability to maintain intestinal organoids. Single-cell RNA sequencing in organoids identified Akt-mTOR (mammalian target of rapamycin) pathway hyperactivation upon iDKO, and pharmacological Akt-mTOR axis inhibition rescued the iDKO defects. Our results demonstrate a requirement for Cbl/Cblb in the maintenance of ISCs by fine-tuning the Akt-mTOR axis to balance stem cell maintenance vs. commitment to differentiation.
Collapse
Affiliation(s)
- Neha Zutshi
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bhopal C. Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Pinaki Mondal
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Wei An
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin T. Goetz
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shuo Wang
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sicong Li
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Matthew D. Storck
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David F. Mercer
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Adrian R. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sarah P. Thayer
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jennifer D. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chi Lin
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Vimla Band
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
189
|
Feng J, Dong H, Lischinsky JE, Zhou J, Deng F, Zhuang C, Miao X, Wang H, Li G, Cai R, Xie H, Cui G, Lin D, Li Y. Monitoring norepinephrine release in vivo using next-generation GRAB NE sensors. Neuron 2024; 112:1930-1942.e6. [PMID: 38547869 PMCID: PMC11364517 DOI: 10.1016/j.neuron.2024.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 01/21/2024] [Accepted: 03/01/2024] [Indexed: 06/22/2024]
Abstract
Norepinephrine (NE) is an essential biogenic monoamine neurotransmitter. The first-generation NE sensor makes in vivo, real-time, cell-type-specific and region-specific NE detection possible, but its low NE sensitivity limits its utility. Here, we developed the second-generation GPCR-activation-based NE sensors (GRABNE2m and GRABNE2h) with a superior response and high sensitivity and selectivity to NE both in vitro and in vivo. Notably, these sensors can detect NE release triggered by either optogenetic or behavioral stimuli in freely moving mice, producing robust signals in the locus coeruleus and hypothalamus. With the development of a novel transgenic mouse line, we recorded both NE release and calcium dynamics with dual-color fiber photometry throughout the sleep-wake cycle; moreover, dual-color mesoscopic imaging revealed cell-type-specific spatiotemporal dynamics of NE and calcium during sensory processing and locomotion. Thus, these new GRABNE sensors are valuable tools for monitoring the precise spatiotemporal release of NE in vivo, providing new insights into the physiological and pathophysiological roles of NE.
Collapse
Affiliation(s)
- Jiesi Feng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.
| | - Hui Dong
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Julieta E Lischinsky
- Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Jingheng Zhou
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Fei Deng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Chaowei Zhuang
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xiaolei Miao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, 100020 Beijing, China
| | - Huan Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Guochuan Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ruyi Cai
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Hao Xie
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Guohong Cui
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Dayu Lin
- Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Chinese Institute for Brain Research, Beijing 102206, China; Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518055, China; National Biomedical Imaging Center, Peking University, Beijing 100871, China.
| |
Collapse
|
190
|
Zerbib L, Ladraa S, Fraissenon A, Bayard C, Firpion M, Venot Q, Protic S, Hoguin C, Thomas A, Fraitag S, Duong JP, Kaltenbach S, Balducci E, Lefevre C, Villarese P, Asnafi V, Broissand C, Goudin N, Nemazanyy I, Autret G, Tavitian B, Legendre C, Arzouk N, Minard-Colin V, Chopinet C, Dussiot M, Adams DM, Mirault T, Guibaud L, Isenring P, Canaud G. Targeted therapy for capillary-venous malformations. Signal Transduct Target Ther 2024; 9:146. [PMID: 38880808 PMCID: PMC11180659 DOI: 10.1038/s41392-024-01862-9] [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: 10/02/2023] [Revised: 04/19/2024] [Accepted: 05/12/2024] [Indexed: 06/18/2024] Open
Abstract
Sporadic venous malformations are genetic conditions primarily caused by somatic gain-of-function mutation of PIK3CA or TEK, an endothelial transmembrane receptor signaling through PIK3CA. Venous malformations are associated with pain, bleedings, thrombosis, pulmonary embolism, esthetic deformities and, in severe cases, life-threatening situations. No authorized medical treatment exists for patients with venous malformations. Here, we created a genetic mouse model of PIK3CA-related capillary venous malformations that replicates patient phenotypes. We showed that these malformations only partially signal through AKT proteins. We compared the efficacy of different drugs, including rapamycin, a mTORC1 inhibitor, miransertib, an AKT inhibitor and alpelisib, a PI3Kα inhibitor at improving the lesions seen in the mouse model. We demonstrated the effectiveness of alpelisib in preventing vascular malformations' occurrence, improving the already established ones, and prolonging survival. Considering these findings, we were authorized to treat 25 patients with alpelisib, including 7 children displaying PIK3CA (n = 16) or TEK (n = 9)-related capillary venous malformations resistant to usual therapies including sirolimus, debulking surgical procedures or percutaneous sclerotherapies. We assessed the volume of vascular malformations using magnetic resonance imaging (MRI) for each patient. Alpelisib demonstrated improvement in all 25 patients. Vascular malformations previously considered intractable were reduced and clinical symptoms were attenuated. MRI showed a decrease of 33.4% and 27.8% in the median volume of PIK3CA and TEK malformations respectively, over 6 months on alpelisib. In conclusion, this study supports PI3Kα inhibition as a promising therapeutic strategy in patients with PIK3CA or TEK-related capillary venous malformations.
Collapse
Affiliation(s)
- Lola Zerbib
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Sophia Ladraa
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Antoine Fraissenon
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, HCL, Bron, France
- CREATIS UMR 5220, Villeurbanne, 69100, France
- Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne, France
| | - Charles Bayard
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Marina Firpion
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Quitterie Venot
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Sanela Protic
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Clément Hoguin
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Amandine Thomas
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Sylvie Fraitag
- Service d'Anatomie pathologique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Jean-Paul Duong
- Université Paris Cité, Paris, France
- Service d'Anatomie pathologique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Sophie Kaltenbach
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Estelle Balducci
- Université Paris Cité, Paris, France
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Coline Lefevre
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Patrick Villarese
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Vahid Asnafi
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | | | - Nicolas Goudin
- Necker Bio-Image Analysis, INSERM US24/CNRS UMS 3633, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS 3633, Paris, France
| | - Gwennhael Autret
- Plateforme Imageries du Vivant, Université de Paris, PARCC, INSERM, Paris, France
| | - Bertrand Tavitian
- Plateforme Imageries du Vivant, Université de Paris, PARCC, INSERM, Paris, France
| | - Christophe Legendre
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service de Néphrologie, Transplantation Adultes, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Nadia Arzouk
- Service de Transplantation, Hôpital La Pitié Salpétrière, AP-HP, Paris, France
| | - Veronique Minard-Colin
- Department of Pediatric and Adolescent Oncology, INSERM 1015, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Caroline Chopinet
- Service de Physiologie & Explorations Fonctionnelles Cardiovasculaires, CHU de Lille, Lille, 59000, France
| | - Michael Dussiot
- INSERM U1163, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Denise M Adams
- Division of Oncology, Comprehensive Vascular Anomalies Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tristan Mirault
- Université Paris Cité, Paris, France
- Service de Médecine Vasculaire, hôpital Européen Georges-Pompidou, Paris, France
| | - Laurent Guibaud
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, HCL, Bron, France
| | - Paul Isenring
- Nephrology Research Group, L'Hôtel-Dieu de Québec Research Center, Department of Medicine, Faculty of Medicine, Laval University, Quebec, QC, G1R2J6, Canada
| | - Guillaume Canaud
- Université Paris Cité, Paris, France.
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France.
- Unité de médecine translationnelle et thérapies ciblées, Hôpital Necker-Enfants Malades, AP-HP, Paris, France.
- CNRS UMR8253, Institut Necker-Enfants Malades, Paris, France.
| |
Collapse
|
191
|
Polsani N, Yung T, Thomas E, Phung-Rojas M, Gupta I, Denker J, Lau K, Feng X, Ibarra B, Hopyan S, Atit RP. Mesenchymal Wnts are required for morphogenetic movements of calvarial osteoblasts during apical expansion. Development 2024; 151:dev202596. [PMID: 38814743 PMCID: PMC11234264 DOI: 10.1242/dev.202596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/13/2024] [Indexed: 06/01/2024]
Abstract
Apical expansion of calvarial osteoblast progenitors from the cranial mesenchyme (CM) above the eye is integral to calvarial growth and enclosure of the brain. The cellular behaviors and signals underlying the morphogenetic process of calvarial expansion are unknown. Time-lapse light-sheet imaging of mouse embryos revealed calvarial progenitors intercalate in 3D in the CM above the eye, and exhibit protrusive and crawling activity more apically. CM cells express non-canonical Wnt/planar cell polarity (PCP) core components and calvarial osteoblasts are bidirectionally polarized. We found non-canonical ligand Wnt5a-/- mutants have less dynamic cell rearrangements and protrusive activity. Loss of CM-restricted Wntless (CM-Wls), a gene required for secretion of all Wnt ligands, led to diminished apical expansion of Osx+ calvarial osteoblasts in the frontal bone primordia in a non-cell autonomous manner without perturbing proliferation or survival. Calvarial osteoblast polarization, progressive cell elongation and enrichment for actin along the baso-apical axis were dependent on CM-Wnts. Thus, CM-Wnts regulate cellular behaviors during calvarial morphogenesis for efficient apical expansion of calvarial osteoblasts. These findings also offer potential insights into the etiologies of calvarial dysplasias.
Collapse
Affiliation(s)
- Nikaya Polsani
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Theodora Yung
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Evan Thomas
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Melissa Phung-Rojas
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Isha Gupta
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Julie Denker
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Kimberly Lau
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xiaotian Feng
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Beatriz Ibarra
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Division of Orthopedics, The Hospital for Sick Children and Departments of Molecular Genetics and Surgery, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Radhika P. Atit
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Dermatology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Genetics and Genome Sciences, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
192
|
Sun Y, Chatterjee S, Lian X, Traylor Z, Sattiraju SR, Xiao Y, Dilliard SA, Sung YC, Kim M, Lee SM, Moore S, Wang X, Zhang D, Wu S, Basak P, Wang J, Liu J, Mann RJ, LePage DF, Jiang W, Abid S, Hennig M, Martinez A, Wustman BA, Lockhart DJ, Jain R, Conlon RA, Drumm ML, Hodges CA, Siegwart DJ. In vivo editing of lung stem cells for durable gene correction in mice. Science 2024; 384:1196-1202. [PMID: 38870301 DOI: 10.1126/science.adk9428] [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: 09/20/2023] [Accepted: 04/17/2024] [Indexed: 06/15/2024]
Abstract
In vivo genome correction holds promise for generating durable disease cures; yet, effective stem cell editing remains challenging. In this work, we demonstrate that optimized lung-targeting lipid nanoparticles (LNPs) enable high levels of genome editing in stem cells, yielding durable responses. Intravenously administered gene-editing LNPs in activatable tdTomato mice achieved >70% lung stem cell editing, sustaining tdTomato expression in >80% of lung epithelial cells for 660 days. Addressing cystic fibrosis (CF), NG-ABE8e messenger RNA (mRNA)-sgR553X LNPs mediated >95% cystic fibrosis transmembrane conductance regulator (CFTR) DNA correction, restored CFTR function in primary patient-derived bronchial epithelial cells equivalent to Trikafta for F508del, corrected intestinal organoids and corrected R553X nonsense mutations in 50% of lung stem cells in CF mice. These findings introduce LNP-enabled tissue stem cell editing for disease-modifying genome correction.
Collapse
Affiliation(s)
- Yehui Sun
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sumanta Chatterjee
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xizhen Lian
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zachary Traylor
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | - Yufen Xiao
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sean A Dilliard
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yun-Chieh Sung
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Minjeong Kim
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sang M Lee
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Stephen Moore
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xu Wang
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Di Zhang
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shiying Wu
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Pratima Basak
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jialu Wang
- ReCode Therapeutics, Menlo Park, CA 94025, USA
| | - Jing Liu
- ReCode Therapeutics, Menlo Park, CA 94025, USA
| | - Rachel J Mann
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - David F LePage
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Weihong Jiang
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Shadaan Abid
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | - Raksha Jain
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ronald A Conlon
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Mitchell L Drumm
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Craig A Hodges
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Daniel J Siegwart
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
193
|
Dawson M, Flores D, Zou L, Anandasenthil S, Mahesh R, Zavala-Romero O, Arora R. Imaging the dynamics of murine uterine contractions in early pregnancy†. Biol Reprod 2024; 110:1175-1190. [PMID: 38713674 PMCID: PMC11180618 DOI: 10.1093/biolre/ioae071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/03/2024] [Accepted: 05/02/2024] [Indexed: 05/09/2024] Open
Abstract
Uterine muscle contractility is essential for reproductive processes including sperm and embryo transport, and during the uterine cycle to remove menstrual effluent. Even still, uterine contractions have primarily been studied in the context of preterm labor. This is partly due to a lack of methods for studying the uterine muscle contractility in the intact organ. Here, we describe an imaging-based method to evaluate mouse uterine contractility of both the longitudinal and circular muscles in the cycling stages and in early pregnancy. By transforming the image-based data into three-dimensional spatiotemporal contractility maps, we calculate waveform characteristics of muscle contractions, including amplitude, frequency, wavelength, and velocity. We report that the native organ is highly contractile during the progesterone-dominant diestrus stage of the cycle when compared to the estrogen-dominant proestrus and estrus stages. We also observed that during the first phase of uterine embryo movement when clustered embryos move toward the middle of the uterine horn, contractions are dynamic and non-uniform between different segments of the uterine horn. In the second phase of embryo movement, contractions are more uniform and rhythmic throughout the uterine horn. Finally, in Lpar3-/- uteri, which display faster embryo movement, we observe global and regional increases in contractility. Our method provides a means to understand the wave characteristics of uterine smooth muscle in response to modulators and in genetic mutants. Better understanding uterine contractility in the early pregnancy stages is critical for the advancement of artificial reproductive technologies and a possibility of modulating embryo movement during clinical embryo transfers.
Collapse
Affiliation(s)
- Madeline Dawson
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, Michigan, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Diana Flores
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, Michigan, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Lisa Zou
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, Michigan, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Shivani Anandasenthil
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, Michigan, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Rohit Mahesh
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, Michigan, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Olmo Zavala-Romero
- Department of Scientific Computing, Florida State University, Tallahassee, Florida, USA
| | - Ripla Arora
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, Michigan, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
194
|
Miyazaki N, Takami S, Uemura M, Oiki H, Takahashi M, Kawashima H, Kanamori Y, Yoshioka T, Kasahara M, Nakazawa A, Higashi M, Yanagida A, Hiramatsu R, Kanai-Azuma M, Fujishiro J, Kanai Y. Impact of gallbladder hypoplasia on hilar hepatic ducts in biliary atresia. COMMUNICATIONS MEDICINE 2024; 4:111. [PMID: 38862768 PMCID: PMC11166647 DOI: 10.1038/s43856-024-00544-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Biliary atresia (BA) is an intractable disease of unknown cause that develops in the neonatal period. It causes jaundice and liver damage due to the destruction of extrahepatic biliary tracts,. We have found that heterozygous knockout mice of the SRY related HMG-box 17 (Sox17) gene, a master regulator of stem/progenitor cells in the gallbladder wall, exhibit a condition like BA. However, the precise contribution of hypoplastic gallbladder wall to the pathogenesis of hepatobiliary disease in Sox17 heterozygous embryos and human BA remains unclear. METHODS We employed cholangiography and histological analyses in the mouse BA model. Furthermore, we conducted a retrospective analysis of human BA. RESULTS We show that gallbladder wall hypoplasia causes abnormal multiple connections between the hilar hepatic bile ducts and the gallbladder-cystic duct in Sox17 heterozygous embryos. These multiple hilar extrahepatic ducts fuse with the developing intrahepatic duct walls and pull them out of the liver parenchyma, resulting in abnormal intrahepatic duct network and severe cholestasis. In human BA with gallbladder wall hypoplasia (i.e., abnormally reduced expression of SOX17), we also identify a strong association between reduced gallbladder width (a morphometric parameter indicating gallbladder wall hypoplasia) and severe liver injury at the time of the Kasai surgery, like the Sox17-mutant mouse model. CONCLUSIONS Together with the close correlation between gallbladder wall hypoplasia and liver damage in both mouse and human cases, these findings provide an insight into the critical role of SOX17-positive gallbladder walls in establishing functional bile duct networks in the hepatic hilus of neonates.
Collapse
Affiliation(s)
- Nanae Miyazaki
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shohei Takami
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Pediatric Surgery, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Mami Uemura
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Center for Experimental Animals, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hironobu Oiki
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Pediatric Surgery, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Surgery, Saitama Children's Medical Center, Saitama, Saitama, Japan
| | - Masataka Takahashi
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development, Setagaya-ku, Tokyo, Japan
| | - Hiroshi Kawashima
- Department of Surgery, Saitama Children's Medical Center, Saitama, Saitama, Japan
| | - Yutaka Kanamori
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development, Setagaya-ku, Tokyo, Japan
| | - Takako Yoshioka
- Department of Pathology, National Center for Child Health and Development, Setagaya-ku, Tokyo, Japan
| | - Mureo Kasahara
- Organ Transplantation Center, National Center for Child Health and Development, Setagaya-ku, Tokyo, Japan
| | - Atsuko Nakazawa
- Department of Clinical Research, Saitama Children's Medical Center, Saitama, Saitama, Japan
| | - Mayumi Higashi
- Department of Pediatric Surgery, Kyoto Prefectural University of Medicine, Kyoto Kamikyo-ku, Kyoto, Japan
| | - Ayaka Yanagida
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Ryuji Hiramatsu
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Masami Kanai-Azuma
- Center for Experimental Animals, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Jun Fujishiro
- Department of Pediatric Surgery, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yoshiakira Kanai
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
195
|
Poscablo DM, Worthington AK, Smith-Berdan S, Rommel MGE, Manso BA, Adili R, Mok L, Reggiardo RE, Cool T, Mogharrab R, Myers J, Dahmen S, Medina P, Beaudin AE, Boyer SW, Holinstat M, Jonsson VD, Forsberg EC. An age-progressive platelet differentiation path from hematopoietic stem cells causes exacerbated thrombosis. Cell 2024; 187:3090-3107.e21. [PMID: 38749423 PMCID: PMC12047039 DOI: 10.1016/j.cell.2024.04.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 02/05/2024] [Accepted: 04/16/2024] [Indexed: 06/09/2024]
Abstract
Platelet dysregulation is drastically increased with advanced age and contributes to making cardiovascular disorders the leading cause of death of elderly humans. Here, we reveal a direct differentiation pathway from hematopoietic stem cells into platelets that is progressively propagated upon aging. Remarkably, the aging-enriched platelet path is decoupled from all other hematopoietic lineages, including erythropoiesis, and operates as an additional layer in parallel with canonical platelet production. This results in two molecularly and functionally distinct populations of megakaryocyte progenitors. The age-induced megakaryocyte progenitors have a profoundly enhanced capacity to engraft, expand, restore, and reconstitute platelets in situ and upon transplantation and produce an additional platelet population in old mice. The two pools of co-existing platelets cause age-related thrombocytosis and dramatically increased thrombosis in vivo. Strikingly, aging-enriched platelets are functionally hyper-reactive compared with the canonical platelet populations. These findings reveal stem cell-based aging as a mechanism for platelet dysregulation and age-induced thrombosis.
Collapse
Affiliation(s)
- Donna M Poscablo
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Atesh K Worthington
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Stephanie Smith-Berdan
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Marcel G E Rommel
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Bryce A Manso
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lydia Mok
- Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Roman E Reggiardo
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Taylor Cool
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Raana Mogharrab
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jenna Myers
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Steven Dahmen
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Paloma Medina
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Anna E Beaudin
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Scott W Boyer
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vanessa D Jonsson
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Applied Mathematics, Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
196
|
Yamaguchi J, Isnard P, Robil N, de la Grange P, Hoguin C, Schmitt A, Hummel A, Megret J, Goudin N, Luka M, Ménager MM, Masson C, Zarhrate M, Bôle-Feysot C, Janiszewska M, Polyak K, Dairou J, Baldassari S, Baulac S, Broissand C, Legendre C, Terzi F, Canaud G. PIK3CA inhibition in models of proliferative glomerulonephritis and lupus nephritis. J Clin Invest 2024; 134:e176402. [PMID: 38842935 PMCID: PMC11290976 DOI: 10.1172/jci176402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 06/04/2024] [Indexed: 08/02/2024] Open
Abstract
Proliferative glomerulonephritis is a severe condition that often leads to kidney failure. There is a significant lack of effective treatment for these disorders. Here, following the identification of a somatic PIK3CA gain-of-function mutation in podocytes of a patient, we demonstrate using multiple genetically engineered mouse models, single-cell RNA sequencing, and spatial transcriptomics the crucial role played by this pathway for proliferative glomerulonephritis development by promoting podocyte proliferation, dedifferentiation, and inflammation. Additionally, we show that alpelisib, a PI3Kα inhibitor, improves glomerular lesions and kidney function in different mouse models of proliferative glomerulonephritis and lupus nephritis by targeting podocytes. Surprisingly, we determined that pharmacological inhibition of PI3Kα affects B and T lymphocyte populations in lupus nephritis mouse models, with a decrease in the production of proinflammatory cytokines, autoantibodies, and glomerular complement deposition, which are all characteristic features of PI3Kδ inhibition, the primary PI3K isoform expressed in lymphocytes. Importantly, PI3Kα inhibition does not impact lymphocyte function under normal conditions. These findings were then confirmed in human lymphocytes isolated from patients with active lupus nephritis. In conclusion, we demonstrate the major role played by PI3Kα in proliferative glomerulonephritis and show that in this condition, alpelisib acts on both podocytes and the immune system.
Collapse
Affiliation(s)
- Junna Yamaguchi
- Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Pierre Isnard
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service d’Anatomie pathologique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Noémie Robil
- Genosplice Technology, Paris Biotech Santé, Paris, France
| | | | - Clément Hoguin
- Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | | | - Aurélie Hummel
- Service de Néphrologie, Transplantation Adultes, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Jérôme Megret
- Structure Fédérative de Recherche Necker, INSERM US24, CNRS UAR 3633, Institut Necker-Enfants Malades, Paris, France
| | - Nicolas Goudin
- Structure Fédérative de Recherche Necker, INSERM US24, CNRS UAR 3633, Institut Necker-Enfants Malades, Paris, France
| | - Marine Luka
- Inflammatory Responses and Transcriptomic Networks in Diseases
- INSERM U1163
| | - Mickaël M. Ménager
- Inflammatory Responses and Transcriptomic Networks in Diseases
- INSERM U1163
| | - Cécile Masson
- Bioinformatics Platform, Structure Fédérative de Recherche Necker, INSERM UMR1163, Université de Paris, and
| | | | | | - Michalina Janiszewska
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technologies, Jupiter, Florida, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Julien Dairou
- Université Paris Cité, Paris, France
- Laboratoire de Chimie et Biologie Pharmacologiques et Toxicologiques, Paris, France
| | - Sara Baldassari
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, Paris, France
| | | | - Christophe Legendre
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service de Néphrologie, Transplantation Adultes, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Fabiola Terzi
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Guillaume Canaud
- Université Paris Cité, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| |
Collapse
|
197
|
Huycke TR, Häkkinen TJ, Miyazaki H, Srivastava V, Barruet E, McGinnis CS, Kalantari A, Cornwall-Scoones J, Vaka D, Zhu Q, Jo H, Oria R, Weaver VM, DeGrado WF, Thomson M, Garikipati K, Boffelli D, Klein OD, Gartner ZJ. Patterning and folding of intestinal villi by active mesenchymal dewetting. Cell 2024; 187:3072-3089.e20. [PMID: 38781967 PMCID: PMC11166531 DOI: 10.1016/j.cell.2024.04.039] [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: 06/12/2023] [Revised: 12/30/2023] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Tissue folds are structural motifs critical to organ function. In the intestine, bending of a flat epithelium into a periodic pattern of folds gives rise to villi, finger-like protrusions that enable nutrient absorption. However, the molecular and mechanical processes driving villus morphogenesis remain unclear. Here, we identify an active mechanical mechanism that simultaneously patterns and folds the intestinal epithelium to initiate villus formation. At the cellular level, we find that PDGFRA+ subepithelial mesenchymal cells generate myosin II-dependent forces sufficient to produce patterned curvature in neighboring tissue interfaces. This symmetry-breaking process requires altered cell and extracellular matrix interactions that are enabled by matrix metalloproteinase-mediated tissue fluidization. Computational models, together with in vitro and in vivo experiments, revealed that these cellular features manifest at the tissue level as differences in interfacial tensions that promote mesenchymal aggregation and interface bending through a process analogous to the active dewetting of a thin liquid film.
Collapse
Affiliation(s)
- Tyler R Huycke
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Teemu J Häkkinen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Hikaru Miyazaki
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Emilie Barruet
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Christopher S McGinnis
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Ali Kalantari
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jake Cornwall-Scoones
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Dedeepya Vaka
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Qin Zhu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Roger Oria
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Krishna Garikipati
- Departments of Mechanical Engineering, and Mathematics, University of Michigan, Ann Arbor, MI, USA
| | - Dario Boffelli
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA.
| | - Zev J Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
198
|
Adkins-Threats M, Arimura S, Huang YZ, Divenko M, To S, Mao H, Zeng Y, Hwang JY, Burclaff JR, Jain S, Mills JC. Metabolic regulator ERRγ governs gastric stem cell differentiation into acid-secreting parietal cells. Cell Stem Cell 2024; 31:886-903.e8. [PMID: 38733994 PMCID: PMC11162331 DOI: 10.1016/j.stem.2024.04.016] [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] [Revised: 02/26/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024]
Abstract
Parietal cells (PCs) produce gastric acid to kill pathogens and aid digestion. Dysregulated PC census is common in disease, yet how PCs differentiate is unclear. Here, we identify the PC progenitors arising from isthmal stem cells, using mouse models and human gastric cells, and show that they preferentially express cell-metabolism regulator and orphan nuclear receptor Estrogen-related receptor gamma (Esrrg, encoding ERRγ). Esrrg expression facilitated the tracking of stepwise molecular, cellular, and ultrastructural stages of PC differentiation. EsrrgP2ACreERT2 lineage tracing revealed that Esrrg expression commits progenitors to differentiate into mature PCs. scRNA-seq indicated the earliest Esrrg+ PC progenitors preferentially express SMAD4 and SP1 transcriptional targets and the GTPases regulating acid-secretion signal transduction. As progenitors matured, ERRγ-dependent metabolic transcripts predominated. Organoid and mouse studies validated the requirement of ERRγ for PC differentiation. Our work chronicles stem cell differentiation along a single lineage in vivo and suggests ERRγ as a therapeutic target for PC-related disorders.
Collapse
Affiliation(s)
- Mahliyah Adkins-Threats
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Division of Biomedical and Biological Sciences, Washington University, St. Louis, MO 63130, USA; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sumimasa Arimura
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yang-Zhe Huang
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Margarita Divenko
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sarah To
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Heather Mao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yongji Zeng
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jenie Y Hwang
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Laboratory Medicine, University of Texas Health San Antonio, San Antonio, TX 78249, USA
| | - Joseph R Burclaff
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - Shilpa Jain
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jason C Mills
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
199
|
Zhu Y, Ackers-Johnson M, Shanmugam MK, Pakkiri LS, Drum CL, Yanpu C, Kim J, Paltzer WG, Mahmoud AI, Wen Tan WL, Lee MCJ, Jianming J, Luu DAT, Ng SL, Li PYQ, Anhui W, Rong Q, Ong GJX, Ng Yu T, Haigh JJ, Tiang Z, Richards AM, Foo R. Asparagine Synthetase Marks a Distinct Dependency Threshold for Cardiomyocyte Dedifferentiation. Circulation 2024; 149:1833-1851. [PMID: 38586957 PMCID: PMC11147732 DOI: 10.1161/circulationaha.123.063965] [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: 03/05/2023] [Accepted: 01/23/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Adult mammalian cardiomyocytes have limited proliferative capacity, but in specifically induced contexts they traverse through cell-cycle reentry, offering the potential for heart regeneration. Endogenous cardiomyocyte proliferation is preceded by cardiomyocyte dedifferentiation (CMDD), wherein adult cardiomyocytes revert to a less matured state that is distinct from the classical myocardial fetal stress gene response associated with heart failure. However, very little is known about CMDD as a defined cardiomyocyte cell state in transition. METHODS Here, we leveraged 2 models of in vitro cultured adult mouse cardiomyocytes and in vivo adeno-associated virus serotype 9 cardiomyocyte-targeted delivery of reprogramming factors (Oct4, Sox2, Klf4, and Myc) in adult mice to study CMDD. We profiled their transcriptomes using RNA sequencing, in combination with multiple published data sets, with the aim of identifying a common denominator for tracking CMDD. RESULTS RNA sequencing and integrated analysis identified Asparagine Synthetase (Asns) as a unique molecular marker gene well correlated with CMDD, required for increased asparagine and also for distinct fluxes in other amino acids. Although Asns overexpression in Oct4, Sox2, Klf4, and Myc cardiomyocytes augmented hallmarks of CMDD, Asns deficiency led to defective regeneration in the neonatal mouse myocardial infarction model, increased cell death of cultured adult cardiomyocytes, and reduced cell cycle in Oct4, Sox2, Klf4, and Myc cardiomyocytes, at least in part through disrupting the mammalian target of rapamycin complex 1 pathway. CONCLUSIONS We discovered a novel gene Asns as both a molecular marker and an essential mediator, marking a distinct threshold that appears in common for at least 4 models of CMDD, and revealing an Asns/mammalian target of rapamycin complex 1 axis dependency for dedifferentiating cardiomyocytes. Further study will be needed to extrapolate and assess its relevance to other cell state transitions as well as in heart regeneration.
Collapse
Affiliation(s)
- Yike Zhu
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Matthew Ackers-Johnson
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Muthu K Shanmugam
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Leroy Sivappiragasam Pakkiri
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Chester Lee Drum
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Chen Yanpu
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Johnny Kim
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhein/Main, Bad Nauheim, Germany
| | - Wyatt G. Paltzer
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ahmed I. Mahmoud
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Wilson Lek Wen Tan
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Mick Chang Jie Lee
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Jiang Jianming
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Danh Anh Tuan Luu
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Shi Ling Ng
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Peter Yi Qing Li
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Wang Anhui
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Centre, Peking University
- State Key Laboratory of Vascular Homeostasis and Remodelling, Peking University
| | - Qi Rong
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Centre, Peking University
- State Key Laboratory of Vascular Homeostasis and Remodelling, Peking University
| | - Gabriel Jing Xiang Ong
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Timothy Ng Yu
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Jody J. Haigh
- CancerCare Manitoba Research Institute, Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- VIB, Flanders Institute for Biotechnology, Ghent University, Ghent, Belgium
| | - Zenia Tiang
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - A. Mark Richards
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
- Christchurch Heart Institute, University of Otago, New Zealand
| | - Roger Foo
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| |
Collapse
|
200
|
Collins BC, Shapiro JB, Scheib MM, Musci RV, Verma M, Kardon G. Three-dimensional imaging studies in mice identify cellular dynamics of skeletal muscle regeneration. Dev Cell 2024; 59:1457-1474.e5. [PMID: 38569550 PMCID: PMC11153043 DOI: 10.1016/j.devcel.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 12/06/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
The function of many organs, including skeletal muscle, depends on their three-dimensional structure. Muscle regeneration therefore requires not only reestablishment of myofibers but also restoration of tissue architecture. Resident muscle stem cells (SCs) are essential for regeneration, but how SCs regenerate muscle architecture is largely unknown. We address this problem using genetic labeling of mouse SCs and whole-mount imaging to reconstruct, in three dimensions, muscle regeneration. Unexpectedly, we found that myofibers form via two distinct phases of fusion and the residual basement membrane of necrotic myofibers is critical for promoting fusion and orienting regenerated myofibers. Furthermore, the centralized myonuclei characteristic of regenerated myofibers are associated with myofibrillogenesis and endure months post injury. Finally, we elucidate two cellular mechanisms for the formation of branched myofibers, a pathology characteristic of diseased muscle. We provide a synthesis of the cellular events of regeneration and show that these differ from those used during development.
Collapse
Affiliation(s)
- Brittany C Collins
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Jacob B Shapiro
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Mya M Scheib
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Robert V Musci
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Mayank Verma
- Department of Pediatrics, Division of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|