1
|
Tso P, Bernier-Latmani J, Petrova TV, Liu M. Transport functions of intestinal lymphatic vessels. Nat Rev Gastroenterol Hepatol 2025; 22:127-145. [PMID: 39496888 DOI: 10.1038/s41575-024-00996-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 11/06/2024]
Abstract
Lymphatic vessels are crucial for fluid absorption and the transport of peripheral immune cells to lymph nodes. However, in the small intestine, the lymphatic fluid is rich in diet-derived lipids incorporated into chylomicrons and gut-specific immune cells. Thus, intestinal lymphatic vessels have evolved to handle these unique cargoes and are critical for systemic dietary lipid delivery and metabolism. This Review covers mechanisms of lipid absorption from epithelial cells to the lymphatics as well as unique features of the gut microenvironment that affect these functions. Moreover, we discuss details of the intestinal lymphatics in gut immune cell trafficking and insights into the role of inter-organ communication. Lastly, we highlight the particularities of fat absorption that can be harnessed for efficient lipid-soluble drug distribution for novel therapies, including the ability of chylomicron-associated drugs to bypass first-pass liver metabolism for systemic delivery. In all, this Review will help to promote an understanding of intestinal lymphatic-systemic interactions to guide future research directions.
Collapse
Affiliation(s)
- Patrick Tso
- Department of Pathology & Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Jeremiah Bernier-Latmani
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Min Liu
- Department of Pathology & Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
2
|
Mohanakrishnan V, Sivaraj KK, Jeong HW, Bovay E, Dharmalingam B, Bixel MG, Dinh VV, Petkova M, Paredes Ugarte I, Kuo YT, Gurusamy M, Raftrey B, Chu NTL, Das S, Rios Coronado PE, Stehling M, Sävendahl L, Chagin AS, Mäkinen T, Red-Horse K, Adams RH. Specialized post-arterial capillaries facilitate adult bone remodelling. Nat Cell Biol 2024; 26:2020-2034. [PMID: 39528700 PMCID: PMC11628402 DOI: 10.1038/s41556-024-01545-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/23/2024] [Indexed: 11/16/2024]
Abstract
The vasculature of the skeletal system is crucial for bone formation, homoeostasis and fracture repair, yet the diversity and specialization of bone-associated vessels remain poorly understood. Here we identify a specialized type of post-arterial capillary, termed type R, involved in bone remodelling. Type R capillaries emerge during adolescence around trabecular bone, possess a distinct morphology and molecular profile, and are associated with osteoprogenitors and bone-resorbing osteoclasts. Endothelial cell-specific overexpression of the transcription factor DACH1 in postnatal mice induces a strong increase in arteries and type R capillaries, leading to local metabolic changes and enabling trabecular bone formation in normally highly hypoxic areas of the diaphysis. Indicating potential clinical relevance of type R capillaries, these vessels respond to anti-osteoporosis treatments and emerge during ageing inside porous structures that are known to weaken compact bone. Our work outlines fundamental principles of vessel specialization in the developing, adult and ageing skeletal system.
Collapse
Affiliation(s)
- Vishal Mohanakrishnan
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Kishor K Sivaraj
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Hyun-Woo Jeong
- Max Planck Institute for Molecular Biomedicine, Single Cell Multi-Omics Laboratory, Münster, Germany
| | - Esther Bovay
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | | | - M Gabriele Bixel
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Van Vuong Dinh
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Milena Petkova
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Isidora Paredes Ugarte
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Yi-Tong Kuo
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Malarvizhi Gurusamy
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Brian Raftrey
- Department of Biology, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Nelson Tsz Long Chu
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Soumyashree Das
- Department of Biology, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India
| | - Pamela E Rios Coronado
- Department of Biology, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Martin Stehling
- Max Planck Institute for Molecular Biomedicine, Flow Cytometry Unit, Münster, Germany
| | - Lars Sävendahl
- Department of Women's and Children's Health, Karolinska Institutet and Pediatric Endocrinology Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Andrei S Chagin
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Taija Mäkinen
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
- Wihuri Research Institute, Helsinki, Finland
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany.
| |
Collapse
|
3
|
Frith ME, Kashyap PC, Linden DR, Theriault B, Chang EB. Microbiota-dependent early-life programming of gastrointestinal motility. iScience 2024; 27:110895. [PMID: 39351201 PMCID: PMC11440258 DOI: 10.1016/j.isci.2024.110895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/17/2023] [Accepted: 09/04/2024] [Indexed: 10/04/2024] Open
Abstract
Gastrointestinal microbes modulate peristalsis and stimulate the enteric nervous system (ENS), whose development, as in the central nervous system (CNS), continues into the murine postweaning period. Given that adult CNS function depends on stimuli received during critical periods of postnatal development, we hypothesized that adult ENS function, namely motility, depends on microbial stimuli during similar critical periods. We gave fecal microbiota transplantation (FMT) to germ-free mice at weaning or as adults and found that only the mice given FMT at weaning recovered normal transit, while those given FMT as adults showed limited improvements. RNA sequencing (RNA-seq) of colonic muscularis propria revealed enrichments in neuron developmental pathways in mice exposed to gut microbes earlier in life, while mice exposed later-or not at all-showed exaggerated expression of inflammatory pathways. These findings highlight a microbiota-dependent sensitive period in ENS development, pointing to potential roles of the early-life microbiome in later-life dysmotility.
Collapse
Affiliation(s)
- Mary E. Frith
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Purna C. Kashyap
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - David R. Linden
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Betty Theriault
- Department of Surgery, University of Chicago, Chicago, IL 60637, USA
| | - Eugene B. Chang
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
4
|
Schrenk S, Bischoff LJ, Boscolo E. Protocol for three-dimensional whole-mount imaging of the vascular network in the intestinal muscle. STAR Protoc 2024; 5:103170. [PMID: 38968077 PMCID: PMC11269289 DOI: 10.1016/j.xpro.2024.103170] [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: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 07/07/2024] Open
Abstract
Three-dimensional (3D) imaging of vascular networks is essential for the investigation of vascular patterning and organization. Here, we present a step-by-step protocol for the 3D visualization of the vasculature within whole-mount preparations of the mouse intestinal muscularis propria layer. We then detail the quantitative analysis of the resulting images for parameters such as vessel density, vessel diameter, the number of endothelial cells, and proliferation. The protocol can be easily extended to study cell-cell interactions such as neuro-vascular or immune-vascular interactions. For complete details on the use and execution of this protocol, please refer to Schrenk et al.1.
Collapse
Affiliation(s)
- Sandra Schrenk
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Lindsay J Bischoff
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Elisa Boscolo
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
5
|
Peter B, Rebeaud J, Vigne S, Bressoud V, Phillips N, Ruiz F, Petrova TV, Bernier-Latmani J, Pot C. Perivascular B cells link intestinal angiogenesis to immunity and to the gut-brain axis during neuroinflammation. J Autoimmun 2024; 148:103292. [PMID: 39067313 DOI: 10.1016/j.jaut.2024.103292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/28/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Disruption of gut barrier function and intestinal immune cell homeostasis are increasingly considered critical players in pathogenesis of extra-intestinal inflammatory diseases, including multiple sclerosis (MS) and its prototypical animal model, the experimental autoimmune encephalomyelitis (EAE). Breakdown of epithelial barriers increases intestinal permeability and systemic dissemination of microbiota-derived molecules. However, whether the gut-vascular barrier (GVB) is altered during EAE has not been reported. Here, we demonstrate that endothelial cell proliferation and vessel permeability increase before EAE clinical onset, leading to vascular remodeling and expansion of intestinal villi capillary bed during disease symptomatic phase in an antigen-independent manner. Concomitant to onset of angiogenesis observed prior to neurological symptoms, we identify an increase of intestinal perivascular immune cells characterized by the surface marker lymphatic vessel endothelial hyaluronic acid receptor 1 (LYVE-1). LYVE-1+ is expressed more frequently on B cells that show high levels of CD73 and have proangiogenic properties. B cell depletion was sufficient to mitigate enteric blood endothelial cell proliferation following immunization for EAE. In conclusion, we propose that altered intestinal vasculature driven by a specialized LYVE-1+ B cell subset promotes angiogenesis and that loss of GVB function is implicated in EAE development and autoimmunity.
Collapse
Affiliation(s)
- Benjamin Peter
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Jessica Rebeaud
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Solenne Vigne
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Valentine Bressoud
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Nicholas Phillips
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Florian Ruiz
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research, Epalinges, 1066, Switzerland
| | - Jeremiah Bernier-Latmani
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research, Epalinges, 1066, Switzerland
| | - Caroline Pot
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland.
| |
Collapse
|
6
|
Wang H, Barry K, Zaini A, Coakley G, Moyat M, Daunt CP, Wickramasinghe LC, Azzoni R, Chatzis R, Yumnam B, Camberis M, Le Gros G, Perdijk O, Foong JPP, Bornstein JC, Marsland BJ, Harris NL. Helminth infection driven gastrointestinal hypermotility is independent of eosinophils and mediated by alterations in smooth muscle instead of enteric neurons. PLoS Pathog 2024; 20:e1011766. [PMID: 39141685 PMCID: PMC11346963 DOI: 10.1371/journal.ppat.1011766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 08/26/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024] Open
Abstract
Intestinal helminth infection triggers a type 2 immune response that promotes a 'weep-and sweep' response characterised by increased mucus secretion and intestinal hypermotility, which function to dislodge the worm from its intestinal habitat. Recent studies have discovered that several other pathogens cause intestinal dysmotility through major alterations to the immune and enteric nervous systems (ENS), and their interactions, within the gastrointestinal tract. However, the involvement of these systems has not been investigated for helminth infections. Eosinophils represent a key cell type recruited by the type 2 immune response and alter intestinal motility under steady-state conditions. Our study aimed to investigate whether altered intestinal motility driven by the murine hookworm, Nippostrongylus brasiliensis, infection involves eosinophils and how the ENS and smooth muscles of the gut are impacted. Eosinophil deficiency did not influence helminth-induced intestinal hypermotility and hypermotility did not involve gross structural or functional changes to the ENS. Hypermotility was instead associated with a dramatic increase in smooth muscle thickness and contractility, an observation that extended to another rodent nematode, Heligmosomoides polygyrus. In summary our data indicate that, in contrast to other pathogens, helminth-induced intestinal hypermotility is driven by largely by myogenic, rather than neurogenic, alterations with such changes occurring independently of eosinophils. (<300 words).
Collapse
Affiliation(s)
- Haozhe Wang
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Kristian Barry
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Aidil Zaini
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Gillian Coakley
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Mati Moyat
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Carmel P. Daunt
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Lakshanie C. Wickramasinghe
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Rossana Azzoni
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Roxanne Chatzis
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Bibek Yumnam
- The Malaghan Institute of Medical Research, Victoria University, Wellington, New Zealand
| | - Mali Camberis
- The Malaghan Institute of Medical Research, Victoria University, Wellington, New Zealand
| | - Graham Le Gros
- The Malaghan Institute of Medical Research, Victoria University, Wellington, New Zealand
| | - Olaf Perdijk
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Jaime P. P. Foong
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Joel C. Bornstein
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Benjamin J. Marsland
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Nicola L. Harris
- Department of Immunology, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Sanketi BD, Mantri M, Huang L, Tavallaei MA, Hu S, Wang MFZ, De Vlaminck I, Kurpios NA. Villus myofibroblasts are developmental and adult progenitors of mammalian gut lymphatic musculature. Dev Cell 2024; 59:1159-1174.e5. [PMID: 38537630 PMCID: PMC11078612 DOI: 10.1016/j.devcel.2024.03.005] [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: 10/27/2023] [Revised: 01/26/2024] [Accepted: 03/01/2024] [Indexed: 05/09/2024]
Abstract
Inside the finger-like intestinal projections called villi, strands of smooth muscle cells contract to propel absorbed dietary fats through the adjacent lymphatic capillary, the lacteal, sending fats into the systemic blood circulation for energy production. Despite this vital function, mechanisms of formation, assembly alongside lacteals, and maintenance of villus smooth muscle are unknown. By combining single-cell RNA sequencing and quantitative lineage tracing of the mouse intestine, we identified a local hierarchy of subepithelial fibroblast progenitors that differentiate into mature smooth muscle fibers via intermediate contractile myofibroblasts. This continuum persists as the major mechanism for villus musculature renewal throughout adult life. The NOTCH3-DLL4 signaling axis governs the assembly of smooth muscle fibers alongside their adjacent lacteals and is required for fat absorption. Our studies identify the ontogeny and maintenance of a poorly defined class of intestinal smooth muscle, with implications for accelerated repair and recovery of digestive function following injury.
Collapse
Affiliation(s)
- Bhargav D Sanketi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Madhav Mantri
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Liqing Huang
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Mohammad A Tavallaei
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Shing Hu
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Michael F Z Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Iwijn De Vlaminck
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA.
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
8
|
Mai H, Luo J, Hoeher L, Al-Maskari R, Horvath I, Chen Y, Kofler F, Piraud M, Paetzold JC, Modamio J, Todorov M, Elsner M, Hellal F, Ertürk A. Whole-body cellular mapping in mouse using standard IgG antibodies. Nat Biotechnol 2024; 42:617-627. [PMID: 37430076 PMCID: PMC11021200 DOI: 10.1038/s41587-023-01846-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 05/26/2023] [Indexed: 07/12/2023]
Abstract
Whole-body imaging techniques play a vital role in exploring the interplay of physiological systems in maintaining health and driving disease. We introduce wildDISCO, a new approach for whole-body immunolabeling, optical clearing and imaging in mice, circumventing the need for transgenic reporter animals or nanobody labeling and so overcoming existing technical limitations. We identified heptakis(2,6-di-O-methyl)-β-cyclodextrin as a potent enhancer of cholesterol extraction and membrane permeabilization, enabling deep, homogeneous penetration of standard antibodies without aggregation. WildDISCO facilitates imaging of peripheral nervous systems, lymphatic vessels and immune cells in whole mice at cellular resolution by labeling diverse endogenous proteins. Additionally, we examined rare proliferating cells and the effects of biological perturbations, as demonstrated in germ-free mice. We applied wildDISCO to map tertiary lymphoid structures in the context of breast cancer, considering both primary tumor and metastases throughout the mouse body. An atlas of high-resolution images showcasing mouse nervous, lymphatic and vascular systems is accessible at http://discotechnologies.org/wildDISCO/atlas/index.php .
Collapse
Affiliation(s)
- Hongcheng Mai
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany
- Munich Medical Research School, Munich, Germany
- Deep Piction GmbH, Munich, Germany
| | - Jie Luo
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany
- Deep Piction GmbH, Munich, Germany
| | - Luciano Hoeher
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
| | - Rami Al-Maskari
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Izabela Horvath
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Ying Chen
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany
- Faculty of Medicine, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Florian Kofler
- Helmholtz Al, Helmholtz Center Munich, Neuherberg, Germany
- Department of Informatics, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marie Piraud
- Helmholtz Al, Helmholtz Center Munich, Neuherberg, Germany
| | - Johannes C Paetzold
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Department of Computing, Imperial College London, London, UK
| | - Jennifer Modamio
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
| | - Mihail Todorov
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Markus Elsner
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
| | - Farida Hellal
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Center Munich, Neuherberg, Germany.
- Institute for Stroke and Dementia Research, Medical Centre of the University of Munich, Ludwig-Maximilians University of Munich, Munich, Germany.
- Deep Piction GmbH, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Graduate School of Neuroscience (GSN), Munich, Germany.
| |
Collapse
|
9
|
Kim G, Chen Z, Li J, Luo J, Castro-Martinez F, Wisniewski J, Cui K, Wang Y, Sun J, Ren X, Crawford SE, Becerra SP, Zhu J, Liu T, Wang S, Zhao K, Wu C. Gut-liver axis calibrates intestinal stem cell fitness. Cell 2024; 187:914-930.e20. [PMID: 38280375 PMCID: PMC10923069 DOI: 10.1016/j.cell.2024.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 10/25/2023] [Accepted: 01/02/2024] [Indexed: 01/29/2024]
Abstract
The gut and liver are recognized to mutually communicate through the biliary tract, portal vein, and systemic circulation. However, it remains unclear how this gut-liver axis regulates intestinal physiology. Through hepatectomy and transcriptomic and proteomic profiling, we identified pigment epithelium-derived factor (PEDF), a liver-derived soluble Wnt inhibitor, which restrains intestinal stem cell (ISC) hyperproliferation to maintain gut homeostasis by suppressing the Wnt/β-catenin signaling pathway. Furthermore, we found that microbial danger signals resulting from intestinal inflammation can be sensed by the liver, leading to the repression of PEDF production through peroxisome proliferator-activated receptor-α (PPARα). This repression liberates ISC proliferation to accelerate tissue repair in the gut. Additionally, treating mice with fenofibrate, a clinical PPARα agonist used for hypolipidemia, enhances colitis susceptibility due to PEDF activity. Therefore, we have identified a distinct role for PEDF in calibrating ISC expansion for intestinal homeostasis through reciprocal interactions between the gut and liver.
Collapse
Affiliation(s)
- Girak Kim
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jian Li
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jialie Luo
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Felipe Castro-Martinez
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jan Wisniewski
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kairong Cui
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yan Wang
- Mass Spectrometry Facility, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jialei Sun
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaobai Ren
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Susan E Crawford
- Department of Surgery, North Shore University Research Institute, University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - S Patricia Becerra
- Section of Protein Structure and Function, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jimin Zhu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Taotao Liu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Sui Wang
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Keji Zhao
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
10
|
Silva TD, Gonçalves-Santos E, Gonçalves RV, Souza RLM, Caetano JE, Caldas IS, Diniz LF, Marques MJ, Novaes RD. Matrix metalloproteinases inhibition reveals the association between inflammation, collagen accumulation and intestinal translocation of Schistosoma mansoni eggs in vivo. Int Immunopharmacol 2024; 127:111353. [PMID: 38086267 DOI: 10.1016/j.intimp.2023.111353] [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: 11/08/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 01/18/2024]
Abstract
Schistosomiasis mansoni is a parasitic infection that causes enterohepatic morbidity associated with severe granulomatous inflammation triggered by parasite eggs. In this disease, granulomatous inflammation leads to intestinal erosion and environmental excretion of S. mansoni eggs from feces, an essential process for propagating the parasite and infecting host organisms. Metalloproteinases (MMP) are involved in S. mansoni-induced hepatic granulomatous inflammation and fibrosis. However, the relationship between MMP and collagen accumulation with the intestinal excretion of parasite eggs remains unclear. Thus, the present study investigated whether MMP inhibition is capable of modulating granulomatous inflammation, collagen accumulation and mechanical resistance to the point of influencing the dynamics between intestinal retention and excretion of S. mansoni eggs in infected mice. Our findings indicated that doxycycline (a potent MMP inhibitor) aggravates intestinal inflammation and subverts collagen dynamics in schistosomiasis. By attenuating MMP-2 and MMP-9 activity, this drug is capable of enhancing fibrosis and mechanical resistance of the intestinal wall, hindering S. mansoni eggs translocation. Although collagen content was not correlated with MMP activity, intestinal retention and fecal excretion of parasite eggs in untreated mice; these correlations were observed for doxycycline-treated animals. Thus, our study provides evidence that doxycycline is able to attenuate fecal elimination of S. mansoni eggs by inhibiting MMP-2 and MMP-9 activity, events potentially associated with excessive collagen accumulation, which increases intestinal mechanical resistance and hinders eggs translocation through the intestinal wall. Variations in intestinal collagen dynamics are relevant since they may represent changes in the environmental dispersion of S. mansoni eggs, bringing repercussions for schistosomiasis propagation.
Collapse
Affiliation(s)
- Thiago D Silva
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil
| | - Elda Gonçalves-Santos
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Departamento de Biologia Animal, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - Raquel L M Souza
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil
| | - José Edson Caetano
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil
| | - Ivo S Caldas
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil
| | - Livia F Diniz
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil
| | - Marcos J Marques
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil
| | - Rômulo D Novaes
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil; Departamento de Biologia Animal, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-000, Minas Gerais, Brazil.
| |
Collapse
|
11
|
Sanketi BD, Mantri M, Huang L, Tavallaei MA, Hu S, Wang MFZ, De Vlaminck I, Kurpios NA. Origin and adult renewal of the gut lacteal musculature from villus myofibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.19.523242. [PMID: 36712064 PMCID: PMC9882374 DOI: 10.1101/2023.01.19.523242] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Intestinal smooth muscles are the workhorse of the digestive system. Inside the millions of finger-like intestinal projections called villi, strands of smooth muscle cells contract to propel absorbed dietary fats through the adjacent lymphatic vessel, called the lacteal, sending fats into the blood circulation for energy production. Despite this vital function, how villus smooth muscles form, how they assemble alongside lacteals, and how they repair throughout life remain unknown. Here we combine single-cell RNA sequencing of the mouse intestine with quantitative lineage tracing to reveal the mechanisms of formation and differentiation of villus smooth muscle cells. Within the highly regenerative villus, we uncover a local hierarchy of subepithelial fibroblast progenitors that progress to become mature smooth muscle fibers, via an intermediate contractile myofibroblast-like phenotype. This continuum persists in the adult intestine as the major source of renewal of villus smooth muscle cells during adult life. We further found that the NOTCH3-DLL4 signaling axis governs the assembly of villus smooth muscles alongside their adjacent lacteal, and we show that this is necessary for gut absorptive function. Overall, our data shed light on the genesis of a poorly defined class of intestinal smooth muscle and pave the way for new opportunities to accelerate recovery of digestive function by stimulating muscle repair.
Collapse
Affiliation(s)
- Bhargav D. Sanketi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University; Ithaca, NY 14853, USA
| | - Madhav Mantri
- Department of Biomedical Engineering, Cornell University; Ithaca, NY 14850, USA
| | - Liqing Huang
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University; Ithaca, NY 14853, USA
| | - Mohammad A. Tavallaei
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University; Ithaca, NY 14853, USA
| | - Shing Hu
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University; Ithaca, NY 14853, USA
| | - Michael F. Z. Wang
- Department of Biomedical Engineering, Cornell University; Ithaca, NY 14850, USA
| | - Iwijn De Vlaminck
- Department of Biomedical Engineering, Cornell University; Ithaca, NY 14850, USA
| | - Natasza A. Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University; Ithaca, NY 14853, USA
| |
Collapse
|
12
|
Bernier-Latmani J, González-Loyola A, Petrova TV. Mechanisms and functions of intestinal vascular specialization. J Exp Med 2024; 221:e20222008. [PMID: 38051275 PMCID: PMC10697212 DOI: 10.1084/jem.20222008] [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: 09/11/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
The intestinal vasculature has been studied for the last 100 years, and its essential role in absorbing and distributing ingested nutrients is well known. Recently, fascinating new insights into the organization, molecular mechanisms, and functions of intestinal vessels have emerged. These include maintenance of intestinal epithelial cell function, coping with microbiota-induced inflammatory pressure, recruiting gut-specific immune cells, and crosstalk with other organs. Intestinal function is also regulated at the systemic and cellular levels, such that the postprandial hyperemic response can direct up to 30% of systemic blood to gut vessels, while micron-sized endothelial cell fenestrations are necessary for nutrient uptake. In this review, we will highlight past discoveries made about intestinal vasculature in the context of new findings of molecular mechanisms underpinning gut function. Such comprehensive understanding of the system will pave the way to breakthroughs in nutrient uptake optimization, drug delivery efficiency, and treatment of human diseases.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | | | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
13
|
Manieri E, Tie G, Malagola E, Seruggia D, Madha S, Maglieri A, Huang K, Fujiwara Y, Zhang K, Orkin SH, Wang TC, He R, McCarthy N, Shivdasani RA. Role of PDGFRA + cells and a CD55 + PDGFRA Lo fraction in the gastric mesenchymal niche. Nat Commun 2023; 14:7978. [PMID: 38042929 PMCID: PMC10693581 DOI: 10.1038/s41467-023-43619-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 11/15/2023] [Indexed: 12/04/2023] Open
Abstract
PDGFRA-expressing mesenchyme supports intestinal stem cells. Stomach epithelia have related niche dependencies, but their enabling mesenchymal cell populations are unknown, in part because previous studies pooled the gastric antrum and corpus. Our high-resolution imaging, transcriptional profiling, and organoid assays identify regional subpopulations and supportive capacities of purified mouse corpus and antral PDGFRA+ cells. Sub-epithelial PDGFRAHi myofibroblasts are principal sources of BMP ligands and two molecularly distinct pools distribute asymmetrically along antral glands but together fail to support epithelial growth in vitro. In contrast, PDGFRALo CD55+ cells strategically positioned beneath gastric glands promote epithelial expansion in the absence of other cells or factors. This population encompasses a small fraction expressing the BMP antagonist Grem1. Although Grem1+ cell ablation in vivo impairs intestinal stem cells, gastric stem cells are spared, implying that CD55+ cell activity in epithelial self-renewal derives from other subpopulations. Our findings shed light on spatial, molecular, and functional organization of gastric mesenchyme and the spectrum of signaling sources for epithelial support.
Collapse
Affiliation(s)
- Elisa Manieri
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Guodong Tie
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
| | - Davide Seruggia
- Department of Hematology, Boston Children's Hospital, Boston, MA, 02115, USA
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Shariq Madha
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Adrianna Maglieri
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Kun Huang
- Molecular Imaging Core and Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Yuko Fujiwara
- Department of Hematology, Boston Children's Hospital, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Kevin Zhang
- Department of Hematology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Stuart H Orkin
- Department of Hematology, Boston Children's Hospital, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
| | - Ruiyang He
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Neil McCarthy
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
14
|
Frith ME, Kashyap PC, Linden DR, Theriault B, Chang EB. Microbiota-dependent early life programming of gastrointestinal motility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566304. [PMID: 38014241 PMCID: PMC10680557 DOI: 10.1101/2023.11.08.566304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Gastrointestinal microbes modulate peristalsis and stimulate the enteric nervous system (ENS), whose development, as in the central nervous system (CNS), continues into the murine postweaning period. Given that adult CNS function depends on stimuli received during critical periods of postnatal development, we hypothesized that adult ENS function, namely motility, depends on microbial stimuli during similar critical periods. We gave fecal microbiota transplantation (FMT) to germ-free mice at weaning or as adults and found that only the mice given FMT at weaning recovered normal transit, while those given FMT as adults showed limited improvements. RNAseq of colonic muscularis propria revealed enrichments in neuron developmental pathways in mice exposed to gut microbes earlier in life, while mice exposed later - or not at all - showed exaggerated expression of inflammatory pathways. These findings highlight a microbiota-dependent sensitive period in ENS development, pointing to potential roles of the early life microbiome in later life dysmotility.
Collapse
|
15
|
Nakai K, Lin H, Yamano S, Tanaka S, Kitamoto S, Saitoh H, Sakuma K, Kurauchi J, Akter E, Konno M, Ishibashi K, Kamata R, Ohashi A, Koseki J, Takahashi H, Yokoyama H, Shiraki Y, Enomoto A, Abe S, Hayakawa Y, Ushiku T, Mutoh M, Fujita Y, Kon S. Wnt activation disturbs cell competition and causes diffuse invasion of transformed cells through NF-κB-MMP21 pathway. Nat Commun 2023; 14:7048. [PMID: 37923722 PMCID: PMC10624923 DOI: 10.1038/s41467-023-42774-6] [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: 12/14/2022] [Accepted: 10/20/2023] [Indexed: 11/06/2023] Open
Abstract
Normal epithelial cells exert their competitive advantage over RasV12-transformed cells and eliminate them into the apical lumen via cell competition. However, the internal or external factors that compromise cell competition and provoke carcinogenesis remain elusive. In this study, we examine the effect of sequential accumulation of gene mutations, mimicking multi-sequential carcinogenesis on RasV12-induced cell competition in intestinal epithelial tissues. Consequently, we find that the directionality of RasV12-cell extrusion in Wnt-activated epithelia is reversed, and transformed cells are delaminated into the basal lamina via non-cell autonomous MMP21 upregulation. Subsequently, diffusively infiltrating, transformed cells develop into highly invasive carcinomas. The elevated production of MMP21 is elicited partly through NF-κB signaling, blockage of which restores apical elimination of RasV12 cells. We further demonstrate that the NF-κB-MMP21 axis is significantly bolstered in early colorectal carcinoma in humans. Collectively, this study shows that cells with high mutational burdens exploit cell competition for their benefit by behaving as unfit cells, endowing them with an invasion advantage.
Collapse
Affiliation(s)
- Kazuki Nakai
- Division of Cancer Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, 278-0022, Japan
| | - Hancheng Lin
- Division of Cancer Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, 278-0022, Japan
| | - Shotaro Yamano
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Kanagawa, 257-0015, Japan
| | - Shinya Tanaka
- Department of Molecular Oncology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Sho Kitamoto
- Division of Microbiology and Immunology, The WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka, 565-0871, Japan
| | - Hitoshi Saitoh
- Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, 277-8577, Japan
| | - Kenta Sakuma
- Division of Cancer Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, 278-0022, Japan
| | - Junpei Kurauchi
- Division of Cancer Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, 278-0022, Japan
| | - Eilma Akter
- Division of Cancer Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, 278-0022, Japan
| | - Masamitsu Konno
- Division of Cancer Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, 278-0022, Japan
| | - Kojiro Ishibashi
- Division of Tumor Cell Biology and Bioimaging, Cancer Research Institute, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Japan
| | - Ryo Kamata
- Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, 277-8577, Japan
| | - Akihiro Ohashi
- Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, 277-8577, Japan
| | - Jun Koseki
- Division of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Hirotaka Takahashi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, 790-8577, Japan
| | - Hideshi Yokoyama
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Yukihiro Shiraki
- Department of Pathology, Nagoya University Hospital, Nagoya, 466-8550, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Hospital, Nagoya, 466-8550, Japan
| | - Sohei Abe
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, 113-8655, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, 113-8655, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, University of Tokyo, Tokyo, 113-8655, Japan
| | - Michihiro Mutoh
- Department of Molecular-Targeting Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yasuyuki Fujita
- Department of Molecular Oncology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Shunsuke Kon
- Division of Cancer Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, 278-0022, Japan.
| |
Collapse
|
16
|
Li Y, Liu S, Zhou K, Wang Y, Chen Y, Hu W, Li S, Li H, Wang Y, Wang Q, He D, Xu H. Neuromedin U programs eosinophils to promote mucosal immunity of the small intestine. Science 2023; 381:1189-1196. [PMID: 37708282 DOI: 10.1126/science.ade4177] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 08/18/2023] [Indexed: 09/16/2023]
Abstract
Eosinophils are granulocytes that play an essential role in type 2 immunity and regulate multiple homeostatic processes in the small intestine (SI). However, the signals that regulate eosinophil activity in the SI at steady state remain poorly understood. Through transcriptome profiling of eosinophils from various mouse tissues, we found that a subset of SI eosinophils expressed neuromedin U (NMU) receptor 1 (NMUR1). Fate-mapping analyses showed that NMUR1 expression in SI eosinophils was programmed by the local microenvironment and further enhanced by inflammation. Genetic perturbation and eosinophil-organoid coculture experiments revealed that NMU-mediated eosinophil activation promotes goblet cell differentiation. Thus, NMU regulates epithelial cell differentiation and barrier immunity by stimulating NMUR1-expressing eosinophils in the SI, which highlights the importance of neuroimmune-epithelial cross-talk in maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Yu Li
- School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Shaorui Liu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Kewen Zhou
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yinsheng Wang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yan Chen
- Center for Inflammatory Bowel Diseases, Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| | - Wen Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| | - Shuyan Li
- Department of Nursing, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| | - Hui Li
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yan Wang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Qiuying Wang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Danyang He
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
| | - Heping Xu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| |
Collapse
|
17
|
Wiggins BG, Wang YF, Burke A, Grunberg N, Vlachaki Walker JM, Dore M, Chahrour C, Pennycook BR, Sanchez-Garrido J, Vernia S, Barr AR, Frankel G, Birdsey GM, Randi AM, Schiering C. Endothelial sensing of AHR ligands regulates intestinal homeostasis. Nature 2023; 621:821-829. [PMID: 37586410 PMCID: PMC10533400 DOI: 10.1038/s41586-023-06508-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
Endothelial cells line the blood and lymphatic vasculature, and act as an essential physical barrier, control nutrient transport, facilitate tissue immunosurveillance and coordinate angiogenesis and lymphangiogenesis1,2. In the intestine, dietary and microbial cues are particularly important in the regulation of organ homeostasis. However, whether enteric endothelial cells actively sense and integrate such signals is currently unknown. Here we show that the aryl hydrocarbon receptor (AHR) acts as a critical node for endothelial cell sensing of dietary metabolites in adult mice and human primary endothelial cells. We first established a comprehensive single-cell endothelial atlas of the mouse small intestine, uncovering the cellular complexity and functional heterogeneity of blood and lymphatic endothelial cells. Analyses of AHR-mediated responses at single-cell resolution identified tissue-protective transcriptional signatures and regulatory networks promoting cellular quiescence and vascular normalcy at steady state. Endothelial AHR deficiency in adult mice resulted in dysregulated inflammatory responses and the initiation of proliferative pathways. Furthermore, endothelial sensing of dietary AHR ligands was required for optimal protection against enteric infection. In human endothelial cells, AHR signalling promoted quiescence and restrained activation by inflammatory mediators. Together, our data provide a comprehensive dissection of the effect of environmental sensing across the spectrum of enteric endothelia, demonstrating that endothelial AHR signalling integrates dietary cues to maintain tissue homeostasis by promoting endothelial cell quiescence and vascular normalcy.
Collapse
Affiliation(s)
- Benjamin G Wiggins
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, London, UK.
| | - Yi-Fang Wang
- MRC London Institute of Medical Sciences, London, UK
| | - Alice Burke
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Nil Grunberg
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Julia M Vlachaki Walker
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Marian Dore
- MRC London Institute of Medical Sciences, London, UK
| | | | - Betheney R Pennycook
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | | | - Santiago Vernia
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Alexis R Barr
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Gad Frankel
- Department of Life Sciences, Imperial College London, London, UK
| | - Graeme M Birdsey
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Anna M Randi
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Chris Schiering
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, London, UK.
| |
Collapse
|
18
|
Tan C, Norden PR, Yu W, Liu T, Ujiie N, Lee SK, Yan X, Dyakiv Y, Aoto K, Ortega S, De Plaen IG, Sampath V, Kume T. Endothelial FOXC1 and FOXC2 promote intestinal regeneration after ischemia-reperfusion injury. EMBO Rep 2023; 24:e56030. [PMID: 37154714 PMCID: PMC10328078 DOI: 10.15252/embr.202256030] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/07/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023] Open
Abstract
Intestinal ischemia underlies several clinical conditions and can result in the loss of the intestinal mucosal barrier. Ischemia-induced damage to the intestinal epithelium is repaired by stimulation of intestinal stem cells (ISCs), and paracrine signaling from the vascular niche regulates intestinal regeneration. Here, we identify FOXC1 and FOXC2 as essential regulators of paracrine signaling in intestinal regeneration after ischemia-reperfusion (I/R) injury. Vascular endothelial cell (EC)- and lymphatic EC (LEC)-specific deletions of Foxc1, Foxc2, or both in mice worsen I/R-induced intestinal damage by causing defects in vascular regrowth, expression of chemokine CXCL12 and Wnt activator R-spondin 3 (RSPO3) in blood ECs (BECs) and LECs, respectively, and activation of Wnt signaling in ISCs. Both FOXC1 and FOXC2 directly bind to regulatory elements of the CXCL12 and RSPO3 loci in BECs and LECs, respectively. Treatment with CXCL12 and RSPO3 rescues the I/R-induced intestinal damage in EC- and LEC-Foxc mutant mice, respectively. This study provides evidence that FOXC1 and FOXC2 are required for intestinal regeneration by stimulating paracrine CXCL12 and Wnt signaling.
Collapse
Affiliation(s)
- Can Tan
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Pieter R Norden
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Wei Yu
- Division of Neonatology, Department of PediatricsChildren's Mercy HospitalKansas CityMOUSA
| | - Ting Liu
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Naoto Ujiie
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Sun Kyong Lee
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Xiaocai Yan
- Department of Pediatrics, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Yaryna Dyakiv
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Kazushi Aoto
- Department of BiochemistryHamamatsu University School of MedicineHamamatsuJapan
| | - Sagrario Ortega
- Mouse Genome Editing Unit, Biotechnology ProgramSpanish National Cancer Research CentreMadridSpain
| | - Isabelle G De Plaen
- Department of Pediatrics, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Venkatesh Sampath
- Division of Neonatology, Department of PediatricsChildren's Mercy HospitalKansas CityMOUSA
| | - Tsutomu Kume
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| |
Collapse
|
19
|
Saygili Demir C, Sabine A, Gong M, Dormond O, Petrova TV. Mechanosensitive mTORC1 signaling maintains lymphatic valves. J Cell Biol 2023; 222:e202207049. [PMID: 37036444 PMCID: PMC10097975 DOI: 10.1083/jcb.202207049] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 01/26/2023] [Accepted: 03/23/2023] [Indexed: 04/11/2023] Open
Abstract
Homeostatic maintenance and repair of lymphatic vessels are essential for health. We investigated the dynamics and the molecular mechanisms of lymphatic endothelial cell (LEC) renewal in adult mesenteric quiescent lymphatic vasculature using label-retention, lineage tracing, and cell ablation strategies. Unlike during development, adult LEC turnover and proliferation was confined to the valve regions of collecting vessels, with valve cells displaying the shortest lifespan. Proliferating valve sinus LECs were the main source for maintenance and repair of lymphatic valves. We identified mechanistic target of rapamycin complex 1 (mTORC1) as a mechanoresponsive pathway activated by fluid shear stress in LECs. Depending on the shear stress level, mTORC1 activity drives division of valve cells or dictates their mechanic resilience through increased protein synthesis. Overactivation of lymphatic mTORC1 in vivo promoted supernumerary valve formation. Our work provides insights into the molecular mechanisms of maintenance of healthy lymphatic vascular system.
Collapse
Affiliation(s)
- Cansaran Saygili Demir
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Amélie Sabine
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Muyun Gong
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Olivier Dormond
- Department of Visceral Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Tatiana V. Petrova
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
20
|
McCarthy N, Tie G, Madha S, He R, Kraiczy J, Maglieri A, Shivdasani RA. Smooth muscle contributes to the development and function of a layered intestinal stem cell niche. Dev Cell 2023; 58:550-564.e6. [PMID: 36924771 PMCID: PMC10089980 DOI: 10.1016/j.devcel.2023.02.012] [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: 06/17/2022] [Revised: 12/05/2022] [Accepted: 02/20/2023] [Indexed: 03/17/2023]
Abstract
Wnt and Rspondin (RSPO) signaling drives proliferation, and bone morphogenetic protein inhibitors (BMPi) impede differentiation, of intestinal stem cells (ISCs). Here, we identify the mouse ISC niche as a complex, multi-layered structure that encompasses distinct mesenchymal and smooth muscle populations. In young and adult mice, diverse sub-cryptal cells provide redundant ISC-supportive factors; few of these are restricted to single cell types. Niche functions refine during postnatal crypt morphogenesis, in part to oppose the dense aggregation of differentiation-promoting BMP+ sub-epithelial myofibroblasts at crypt-villus junctions. Muscularis mucosae, a specialized muscle layer, first appears during this period and supplements neighboring RSPO and BMPi sources. Components of this developing niche are conserved in human fetuses. The in vivo ablation of mouse postnatal smooth muscle increases BMP signaling activity, potently limiting a pre-weaning burst of crypt fission. Thus, distinct and progressively specialized mesenchymal cells together create the milieu that is required to propagate crypts during rapid organ growth and to sustain adult ISCs.
Collapse
Affiliation(s)
- Neil McCarthy
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| | - Guodong Tie
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Shariq Madha
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ruiyang He
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Judith Kraiczy
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Adrianna Maglieri
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA.
| |
Collapse
|
21
|
Valdés Zayas A, Kumari N, Liu K, Neill D, Delahoussaye A, Gonçalves Jorge P, Geyer R, Lin SH, Bailat C, Bochud F, Moeckli R, Koong AC, Bourhis J, Taniguchi CM, Herrera FG, Schüler E. Independent Reproduction of the FLASH Effect on the Gastrointestinal Tract: A Multi-Institutional Comparative Study. Cancers (Basel) 2023; 15:cancers15072121. [PMID: 37046782 PMCID: PMC10093322 DOI: 10.3390/cancers15072121] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
FLASH radiation therapy (RT) is a promising new paradigm in radiation oncology. However, a major question that remains is the robustness and reproducibility of the FLASH effect when different irradiators are used on animals or patients with different genetic backgrounds, diets, and microbiomes, all of which can influence the effects of radiation on normal tissues. To address questions of rigor and reproducibility across different centers, we analyzed independent data sets from The University of Texas MD Anderson Cancer Center and from Lausanne University (CHUV). Both centers investigated acute effects after total abdominal irradiation to C57BL/6 animals delivered by the FLASH Mobetron system. The two centers used similar beam parameters but otherwise conducted the studies independently. The FLASH-enabled animal survival and intestinal crypt regeneration after irradiation were comparable between the two centers. These findings, together with previously published data using a converted linear accelerator, show that a robust and reproducible FLASH effect can be induced as long as the same set of irradiation parameters are used.
Collapse
Affiliation(s)
- Anet Valdés Zayas
- Radio-Oncology Department, AGORA Cancer Research Institute, Lausanne University Hospital, Lausanne University, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - Neeraj Kumari
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kevin Liu
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas, Houston, TX 77030, USA
| | - Denae Neill
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Abagail Delahoussaye
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Patrik Gonçalves Jorge
- Institute of Radiation Physics, Lausanne University Hospital, Lausanne University, Rue du Grand-Pré-1, CH-1007 Lausanne, Switzerland
| | - Reiner Geyer
- Institute of Radiation Physics, Lausanne University Hospital, Lausanne University, Rue du Grand-Pré-1, CH-1007 Lausanne, Switzerland
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas, Houston, TX 77030, USA
| | - Claude Bailat
- Institute of Radiation Physics, Lausanne University Hospital, Lausanne University, Rue du Grand-Pré-1, CH-1007 Lausanne, Switzerland
| | - François Bochud
- Institute of Radiation Physics, Lausanne University Hospital, Lausanne University, Rue du Grand-Pré-1, CH-1007 Lausanne, Switzerland
| | - Raphael Moeckli
- Institute of Radiation Physics, Lausanne University Hospital, Lausanne University, Rue du Grand-Pré-1, CH-1007 Lausanne, Switzerland
| | - Albert C. Koong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas, Houston, TX 77030, USA
| | - Jean Bourhis
- Radio-Oncology Department, AGORA Cancer Research Institute, Lausanne University Hospital, Lausanne University, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - Cullen M. Taniguchi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas, Houston, TX 77030, USA
| | - Fernanda G. Herrera
- Radio-Oncology Department, AGORA Cancer Research Institute, Lausanne University Hospital, Lausanne University, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - Emil Schüler
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas, Houston, TX 77030, USA
| |
Collapse
|
22
|
Manieri E, Tie G, Seruggia D, Madha S, Maglieri A, Huang K, Fujiwara Y, Zhang K, Orkin SH, He R, McCarthy N, Shivdasani RA. Defining the structure, signals, and cellular elements of the gastric mesenchymal niche. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.11.527728. [PMID: 36798304 PMCID: PMC9934611 DOI: 10.1101/2023.02.11.527728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
PDGFRA-expressing mesenchyme provides a niche for intestinal stem cells. Corresponding compartments are unknown in the stomach, where corpus and antral glandular epithelia have similar niche dependencies but are structurally distinct from the intestine and from each other. Previous studies considered antrum and corpus as a whole and did not assess niche functions. Using high-resolution imaging and sequencing, we identify regional subpopulations and niche properties of purified mouse corpus and antral PDGFRA + cells. PDGFRA Hi sub-epithelial myofibroblasts are principal sources of BMP ligands in both gastric segments; two molecularly distinct groups distribute asymmetrically along antral glands but together fail to support epithelial organoids in vitro . In contrast, strategically positioned PDGFRA Lo cells that express CD55 enable corpus and antral organoid growth in the absence of other cellular or soluble factors. Our study provides detailed insights into spatial, molecular, and functional organization of gastric mesenchyme and the spectrum of signaling sources for stem cell support.
Collapse
|
23
|
Mueller JPJ, Dobosz M, O’Brien N, Abdoush N, Giusti AM, Lechmann M, Osl F, Wolf AK, Arellano-Viera E, Shaikh H, Sauer M, Rosenwald A, Herting F, Umaña P, Colombetti S, Pöschinger T, Beilhack A. ROCKETS - a novel one-for-all toolbox for light sheet microscopy in drug discovery. Front Immunol 2023; 14:1034032. [PMID: 36845124 PMCID: PMC9945347 DOI: 10.3389/fimmu.2023.1034032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/06/2023] [Indexed: 02/10/2023] Open
Abstract
Advancing novel immunotherapy strategies requires refined tools in preclinical research to thoroughly assess drug targets, biodistribution, safety, and efficacy. Light sheet fluorescence microscopy (LSFM) offers unprecedented fast volumetric ex vivo imaging of large tissue samples in high resolution. Yet, to date laborious and unstandardized tissue processing procedures have limited throughput and broader applications in immunological research. Therefore, we developed a simple and harmonized protocol for processing, clearing and imaging of all mouse organs and even entire mouse bodies. Applying this Rapid Optical Clearing Kit for Enhanced Tissue Scanning (ROCKETS) in combination with LSFM allowed us to comprehensively study the in vivo biodistribution of an antibody targeting Epithelial Cell Adhesion Molecule (EpCAM) in 3D. Quantitative high-resolution scans of whole organs did not only reveal known EpCAM expression patterns but, importantly, uncovered several new EpCAM-binding sites. We identified gustatory papillae of the tongue, choroid plexi in the brain and duodenal papillae as previously unanticipated locations of high EpCAM expression. Subsequently, we confirmed high EpCAM expression also in human tongue and duodenal specimens. Choroid plexi and duodenal papillae may be considered as particularly sensitive sites due to their importance for liquor production or as critical junctions draining bile and digestive pancreatic enzymes into the small bowel, respectively. These newly gained insights appear highly relevant for clinical translation of EpCAM-addressing immunotherapies. Thus, ROCKETS in combination with LSFM may help to set new standards for preclinical evaluation of immunotherapeutic strategies. In conclusion, we propose ROCKETS as an ideal platform for a broader application of LSFM in immunological research optimally suited for quantitative co-localization studies of immunotherapeutic drugs and defined cell populations in the microanatomical context of organs or even whole mice.
Collapse
Affiliation(s)
- Joerg P. J. Mueller
- Interdisciplinary Center for Clinical Research Laboratory (IZKF) Würzburg, Department of Internal Medicine II, Center for Experimental Molecular Medicine, Würzburg University Hospital, Würzburg, Germany
- Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Michael Dobosz
- Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Nils O’Brien
- Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Nassri Abdoush
- Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Anna Maria Giusti
- Roche Pharmaceutical Research and Early Development, Roche Glycart AG, Schlieren, Switzerland
| | - Martin Lechmann
- Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Franz Osl
- Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Ann-Katrin Wolf
- Interdisciplinary Center for Clinical Research Laboratory (IZKF) Würzburg, Department of Internal Medicine II, Center for Experimental Molecular Medicine, Würzburg University Hospital, Würzburg, Germany
- Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Estibaliz Arellano-Viera
- Interdisciplinary Center for Clinical Research Laboratory (IZKF) Würzburg, Department of Internal Medicine II, Center for Experimental Molecular Medicine, Würzburg University Hospital, Würzburg, Germany
| | - Haroon Shaikh
- Interdisciplinary Center for Clinical Research Laboratory (IZKF) Würzburg, Department of Internal Medicine II, Center for Experimental Molecular Medicine, Würzburg University Hospital, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | | | - Frank Herting
- Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Pablo Umaña
- Roche Pharmaceutical Research and Early Development, Roche Glycart AG, Schlieren, Switzerland
| | - Sara Colombetti
- Roche Pharmaceutical Research and Early Development, Roche Glycart AG, Schlieren, Switzerland
| | - Thomas Pöschinger
- Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Andreas Beilhack
- Interdisciplinary Center for Clinical Research Laboratory (IZKF) Würzburg, Department of Internal Medicine II, Center for Experimental Molecular Medicine, Würzburg University Hospital, Würzburg, Germany
| |
Collapse
|
24
|
van Ineveld RL, Collot R, Román MB, Pagliaro A, Bessler N, Ariese HCR, Kleinnijenhuis M, Kool M, Alieva M, Chuva de Sousa Lopes SM, Wehrens EJ, Rios AC. Multispectral confocal 3D imaging of intact healthy and tumor tissue using mLSR-3D. Nat Protoc 2022; 17:3028-3055. [PMID: 36180532 DOI: 10.1038/s41596-022-00739-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 06/16/2022] [Indexed: 11/09/2022]
Abstract
Revealing the 3D composition of intact tissue specimens is essential for understanding cell and organ biology in health and disease. State-of-the-art 3D microscopy techniques aim to capture tissue volumes on an ever-increasing scale, while also retaining sufficient resolution for single-cell analysis. Furthermore, spatial profiling through multi-marker imaging is fast developing, providing more context and better distinction between cell types. Following these lines of technological advance, we here present a protocol based on FUnGI (fructose, urea and glycerol clearing solution for imaging) optical clearing of tissue before multispectral large-scale single-cell resolution 3D (mLSR-3D) imaging, which implements 'on-the-fly' linear unmixing of up to eight fluorophores during a single acquisition. Our protocol removes the need for repetitive illumination, thereby allowing larger volumes to be scanned with better image quality in less time, also reducing photo-bleaching and file size. To aid in the design of multiplex antibody panels, we provide a fast and manageable intensity equalization assay with automated analysis to design a combination of markers with balanced intensities suitable for mLSR-3D. We demonstrate effective mLSR-3D imaging of various tissues, including patient-derived organoids and xenografted tumors, and, furthermore, describe an optimized workflow for mLSR-3D imaging of formalin-fixed paraffin-embedded samples. Finally, we provide essential steps for 3D image data processing, including shading correction that does not require pre-acquired shading references and 3D inhomogeneity correction to correct fluorescence artefacts often afflicting 3D datasets. Together, this provides a one-week protocol for eight-fluorescent-marker 3D visualization and exploration of intact tissue of various origins at single-cell resolution.
Collapse
Affiliation(s)
- Ravian L van Ineveld
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Raphaël Collot
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Mario Barrera Román
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Anna Pagliaro
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Nils Bessler
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Hendrikus C R Ariese
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Michiel Kleinnijenhuis
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Marcel Kool
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center DKFZ and German Cancer Consortium DKTK, Heidelberg, Germany
| | - Maria Alieva
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | | | - Ellen J Wehrens
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Anne C Rios
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
- Oncode Institute, Utrecht, the Netherlands.
| |
Collapse
|
25
|
Cifarelli V, Peche VS, Abumrad NA. Vascular and lymphatic regulation of gastrointestinal function and disease risk. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159207. [PMID: 35882297 PMCID: PMC9642046 DOI: 10.1016/j.bbalip.2022.159207] [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: 01/17/2022] [Revised: 06/17/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022]
Abstract
The vascular and lymphatic systems in the gut regulate lipid transport while restricting transfer of commensal gut microbiota and directing immune cell trafficking. Increased permeability of the endothelial systems in the intestine associates with passage of antigens and microbiota from the gut into the bloodstream leading to tissue inflammation, the release of pro-inflammatory mediators and ultimately to abnormalities of systemic metabolism. Recent studies show that lipid metabolism maintains homeostasis and function of intestinal blood and lymphatic endothelial cells, BECs and LECs, respectively. This review highlights recent progress in this area, and information related to the contribution of the lipid transporter CD36, abundant in BECs and LECs, to gastrointestinal barrier integrity, inflammation, and to gut regulation of whole body metabolism. The potential role of endothelial lipid delivery in epithelial tissue renewal after injury and consequently in the risk of gastric and intestinal diseases is also discussed.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, USA.
| | - Vivek S Peche
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nada A Abumrad
- Center for Human Nutrition, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
26
|
Hu S, Sevier CS, Kurpios NA. Protocol to detect smooth muscle actin-alpha and measure oxidative damage in neonatal mouse intestine. STAR Protoc 2022; 3:101524. [PMID: 35810413 PMCID: PMC9284457 DOI: 10.1016/j.xpro.2022.101524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/08/2022] [Accepted: 06/13/2022] [Indexed: 11/21/2022] Open
Abstract
This protocol describes how to characterize α-Smooth muscle actin (αSMA) spatiotemporal expression during mouse small intestinal development. Specific tissue fixation preserves αSMA arrangement in low αSMA expressing cells that are conventionally undetectable under αSMA immunofluorescent stain due to inappropriate fixative-caused artificial actin depolymerization. Parallel analysis of αSMA carbonylation allows estimation of oxidative damage in gut muscular lineage. This approach improves the molecular specificity offered by commercialized kits that estimate total protein carbonyl level in cell lysates without protein specificity. For complete details on the use and execution of this protocol, please refer to Hu et al. (2021). Optimal fixative improves αSMA immunofluorescent staining in the mouse intestine Improved cellular resolution of αSMA protein in whole mount immunofluorescent staining Semiquantitative measurement of oxidative damage on specific tissue proteins in vivo This protocol identifies the size distribution of total protein carbonyl in animal tissue
Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
Collapse
|
27
|
Lymphangiocrine signals are required for proper intestinal repair after cytotoxic injury. Cell Stem Cell 2022; 29:1262-1272.e5. [PMID: 35931034 PMCID: PMC9387209 DOI: 10.1016/j.stem.2022.07.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/26/2022] [Accepted: 07/15/2022] [Indexed: 12/20/2022]
Abstract
The intestinal epithelium undergoes continuous renewal and has an exceptional capacity to regenerate after injury. Maintenance and proliferation of intestinal stem cells (ISCs) are regulated by their surrounding niche, largely through Wnt signaling. However, it remains unclear which niche cells produce signals during different injury states, and the role of endothelial cells (ECs) as a component of the ISC niche during homeostasis and after injury has been underappreciated. Here, we show that lymphatic endothelial cells (LECs) reside in proximity to crypt epithelial cells and secrete molecules that support epithelial renewal and repair. LECs are an essential source of Wnt signaling in the small intestine, as loss of LEC-derived Rspo3 leads to a lower number of stem and progenitor cells and hinders recovery after cytotoxic injury. Together, our findings identify LECs as an essential niche component for optimal intestinal recovery after cytotoxic injury.
Collapse
|
28
|
Bernier-Latmani J, Mauri C, Marcone R, Renevey F, Durot S, He L, Vanlandewijck M, Maclachlan C, Davanture S, Zamboni N, Knott GW, Luther SA, Betsholtz C, Delorenzi M, Brisken C, Petrova TV. ADAMTS18 + villus tip telocytes maintain a polarized VEGFA signaling domain and fenestrations in nutrient-absorbing intestinal blood vessels. Nat Commun 2022; 13:3983. [PMID: 35810168 PMCID: PMC9271081 DOI: 10.1038/s41467-022-31571-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 06/21/2022] [Indexed: 12/17/2022] Open
Abstract
The small intestinal villus tip is the first point of contact for lumen-derived substances including nutrients and microbial products. Electron microscopy studies from the early 1970s uncovered unusual spatial organization of small intestinal villus tip blood vessels: their exterior, epithelial-facing side is fenestrated, while the side facing the villus stroma is non-fenestrated, covered by pericytes and harbors endothelial nuclei. Such organization optimizes the absorption process, however the molecular mechanisms maintaining this highly specialized structure remain unclear. Here we report that perivascular LGR5+ villus tip telocytes (VTTs) are necessary for maintenance of villus tip endothelial cell polarization and fenestration by sequestering VEGFA signaling. Mechanistically, unique VTT expression of the protease ADAMTS18 is necessary for VEGFA signaling sequestration through limiting fibronectin accumulation. Therefore, we propose a model in which LGR5+ ADAMTS18+ telocytes are necessary to maintain a “just-right” level and location of VEGFA signaling in intestinal villus blood vasculature to ensure on one hand the presence of sufficient endothelial fenestrae, while avoiding excessive leakiness of the vessels and destabilization of villus tip epithelial structures. The molecular mechanisms ensuring the specialized structure of small intestinal villus tip blood vessels are incompletely understood. Here the authors show that ADAMTS18+ telocytes maintain a “just-right” level and location of VEGFA signaling on intestinal villus blood vessels, thereby ensuring the presence of endothelial fenestrae for nutrient absorption, while avoiding excessive leakiness and destabilization of villus tip epithelial structures.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.
| | - Cristina Mauri
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Rachel Marcone
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - François Renevey
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland
| | - Stephan Durot
- Institute of Molecular Systems Biology ETH, Zurich, Switzerland
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Michael Vanlandewijck
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Department of Medicine-Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Catherine Maclachlan
- Bio Electron Microscopy Laboratory, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Suzel Davanture
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Nicola Zamboni
- Institute of Molecular Systems Biology ETH, Zurich, Switzerland
| | - Graham W Knott
- Bio Electron Microscopy Laboratory, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Sanjiv A Luther
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Department of Medicine-Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Mauro Delorenzi
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.,Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Cathrin Brisken
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland. .,Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland.
| |
Collapse
|
29
|
Ignacio A, Shah K, Bernier-Latmani J, Köller Y, Coakley G, Moyat M, Hamelin R, Armand F, Wong NC, Ramay H, Thomson CA, Burkhard R, Wang H, Dufour A, Geuking MB, McDonald B, Petrova TV, Harris NL, McCoy KD. Small intestinal resident eosinophils maintain gut homeostasis following microbial colonization. Immunity 2022; 55:1250-1267.e12. [PMID: 35709757 DOI: 10.1016/j.immuni.2022.05.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/29/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022]
Abstract
The intestine harbors a large population of resident eosinophils, yet the function of intestinal eosinophils has not been explored. Flow cytometry and whole-mount imaging identified eosinophils residing in the lamina propria along the length of the intestine prior to postnatal microbial colonization. Microscopy, transcriptomic analysis, and mass spectrometry of intestinal tissue revealed villus blunting, altered extracellular matrix, decreased epithelial cell turnover, increased gastrointestinal motility, and decreased lipid absorption in eosinophil-deficient mice. Mechanistically, intestinal epithelial cells released IL-33 in a microbiota-dependent manner, which led to eosinophil activation. The colonization of germ-free mice demonstrated that eosinophil activation in response to microbes regulated villous size alterations, macrophage maturation, epithelial barrier integrity, and intestinal transit. Collectively, our findings demonstrate a critical role for eosinophils in facilitating the mutualistic interactions between the host and microbiota and provide a rationale for the functional significance of their early life recruitment in the small intestine.
Collapse
Affiliation(s)
- Aline Ignacio
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Cumming School of Medicine, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Kathleen Shah
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Jeremiah Bernier-Latmani
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland
| | - Yasmin Köller
- Maurice Müller Laboratories, Department of Biomedical Research, Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
| | - Gillian Coakley
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Mati Moyat
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland; Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Romain Hamelin
- Proteomics Core Facility, Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland
| | - Florence Armand
- Proteomics Core Facility, Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland
| | - Nick C Wong
- Monash Bioinformatics Platform, Monash University, Clayton, VIC 3168, Australia
| | - Hena Ramay
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Carolyn A Thomson
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Cumming School of Medicine, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Regula Burkhard
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute of Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Haozhe Wang
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Antoine Dufour
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Cumming School of Medicine, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Markus B Geuking
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute of Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Braedon McDonald
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4A1, Canada
| | - Tatiana V Petrova
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland; Swiss Institute for Experimental Cancer Research, School of Life Sciences, Swiss Federal Institute of Technology Lausanne, Route Cantonale, 1015 Lausanne, Switzerland
| | - Nicola L Harris
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, 1015 Lausanne, Switzerland; Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia.
| | - Kathy D McCoy
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Cumming School of Medicine, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
30
|
Apelin drives endothelial cell migration toward intestinal progenitor cells. NATURE CARDIOVASCULAR RESEARCH 2022; 1:546-547. [PMID: 39195862 DOI: 10.1038/s44161-022-00079-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
|
31
|
Tisch N, Mogler C, Stojanovic A, Luck R, Korhonen EA, Ellerkmann A, Adler H, Singhal M, Schermann G, Erkert L, Patankar JV, Karakatsani A, Scherr AL, Fuchs Y, Cerwenka A, Wirtz S, Köhler BC, Augustin HG, Becker C, Schmidt T, Ruiz de Almodóvar C. Caspase-8 in endothelial cells maintains gut homeostasis and prevents small bowel inflammation in mice. EMBO Mol Med 2022; 14:e14121. [PMID: 35491615 PMCID: PMC9174885 DOI: 10.15252/emmm.202114121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 12/18/2022] Open
Abstract
The gut has a specific vascular barrier that controls trafficking of antigens and microbiota into the bloodstream. However, the molecular mechanisms regulating the maintenance of this vascular barrier remain elusive. Here, we identified Caspase-8 as a pro-survival factor in mature intestinal endothelial cells that is required to actively maintain vascular homeostasis in the small intestine in an organ-specific manner. In particular, we find that deletion of Caspase-8 in endothelial cells results in small intestinal hemorrhages and bowel inflammation, while all other organs remained unaffected. We also show that Caspase-8 seems to be particularly needed in lymphatic endothelial cells to maintain gut homeostasis. Our work demonstrates that endothelial cell dysfunction, leading to the breakdown of the gut-vascular barrier, is an active driver of chronic small intestinal inflammation, highlighting the role of the intestinal vasculature as a safeguard of organ function.
Collapse
Affiliation(s)
- Nathalie Tisch
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carolin Mogler
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Ana Stojanovic
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Robert Luck
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Emilia A Korhonen
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander Ellerkmann
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of General, Visceral and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Heike Adler
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Mahak Singhal
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Géza Schermann
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Erkert
- Department of Medicine 1, Friedrich-Alexander-University, Erlangen, Germany
| | - Jay V Patankar
- Department of Medicine 1, Friedrich-Alexander-University, Erlangen, Germany
| | - Andromachi Karakatsani
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anna-Lena Scherr
- National Center for Tumor Diseases, Department of Medical Oncology, Internal Medicine VI, Heidelberg University Hospital, Heidelberg, Germany
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology & Regenerative Medicine, Department of Biology, Technion -Israel Institute of Technology, Haifa, Israel
| | - Adelheid Cerwenka
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Immunobiochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Friedrich-Alexander-University, Erlangen, Germany
| | - Bruno Christian Köhler
- National Center for Tumor Diseases, Department of Medical Oncology, Internal Medicine VI, Heidelberg University Hospital, Heidelberg, Germany
| | - Hellmut G Augustin
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-University, Erlangen, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, Heidelberg University, Heidelberg, Germany.,Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine with University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Carmen Ruiz de Almodóvar
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
32
|
Bernier-Latmani J, Cisarovsky C, Mahfoud S, Ragusa S, Dupanloup I, Barras D, Renevey F, Nassiri S, Anderle P, Squadrito ML, Siegert S, Davanture S, González-Loyola A, Fournier N, Luther SA, Benedito R, Valet P, Zhou B, De Palma M, Delorenzi M, Sempoux C, Petrova TV. Apelin-driven endothelial cell migration sustains intestinal progenitor cells and tumor growth. NATURE CARDIOVASCULAR RESEARCH 2022; 1:476-490. [PMID: 35602406 PMCID: PMC7612746 DOI: 10.1038/s44161-022-00061-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
Stem and progenitor cells residing in the intestinal crypts drive the majority of colorectal cancers (CRCs), yet vascular contribution to this niche remains largely unexplored. VEGFA is a key driver of physiological and tumor angiogenesis. Accordingly, current anti-angiogenic cancer therapies target the VEGFA pathway. Here we report that in CRC expansion of the stem/progenitor pool in intestinal crypts requires VEGFA-independent growth and remodeling of blood vessels. Epithelial transformation induced expression of the endothelial peptide apelin, directs migration of distant venous endothelial cells towards progenitor niche vessels ensuring optimal perfusion. In the absence of apelin, loss of injury-inducible PROX1+ epithelial progenitors inhibited both incipient and advanced intestinal tumor growth. Our results establish fundamental principles for the reciprocal communication between vasculature and the intestinal progenitor niche and provide a mechanism for resistance to VEGFA-targeting drugs in CRCs.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Christophe Cisarovsky
- Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Samantha Mahfoud
- Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Simone Ragusa
- Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Isabelle Dupanloup
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - David Barras
- Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - François Renevey
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Sina Nassiri
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Pascale Anderle
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Mario Leonardo Squadrito
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Stefanie Siegert
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Suzel Davanture
- Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Alejandra González-Loyola
- Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Nadine Fournier
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Sanjiv A. Luther
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Rui Benedito
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Philippe Valet
- Institut RESTORE, UMR 1301-INSERM, 5070-CNRS, Université Paul Sabatier, Université de Toulouse, Toulouse, France
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Mauro Delorenzi
- Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Christine Sempoux
- Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Tatiana V. Petrova
- Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland
- Corresponding author.
| |
Collapse
|
33
|
Yan X, Managlia E, Zhao YY, Tan XD, De Plaen IG. Macrophage-derived IGF-1 protects the neonatal intestine against necrotizing enterocolitis by promoting microvascular development. Commun Biol 2022; 5:320. [PMID: 35388142 PMCID: PMC8987083 DOI: 10.1038/s42003-022-03252-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 03/11/2022] [Indexed: 02/07/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a deadly bowel necrotic disease of premature infants. Low levels of plasma IGF-1 predispose premature infants to NEC. While increasing evidence suggests that defective perinatal intestinal microvascular development plays a role in NEC, the involved mechanism remains incompletely understood. We report here that serum and intestinal IGF-1 are developmentally regulated during the perinatal period in mice and decrease during experimental NEC. Neonatal intestinal macrophages produce IGF-1 and promote endothelial cell sprouting in vitro via IGF-1 signaling. In vivo, in the neonatal intestine, macrophage-derived IGF-1 promotes VEGF expression and endothelial cell proliferation and protects against experimental NEC. Exogenous IGF-1 preserves intestinal microvascular density and protects against experimental NEC. In human NEC tissues, villous endothelial cell proliferation and IGF-1- producing macrophages are decreased compared to controls. Together, our results suggest that defective IGF-1-production by neonatal macrophages impairs neonatal intestinal microvascular development and predisposes the intestine to necrotizing enterocolitis.
Collapse
Affiliation(s)
- Xiaocai Yan
- grid.16753.360000 0001 2299 3507Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL USA ,grid.16753.360000 0001 2299 3507Center for Intestinal and Liver Inflammation Research, Stanley Manne Children’s. Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Elizabeth Managlia
- grid.16753.360000 0001 2299 3507Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL USA ,grid.16753.360000 0001 2299 3507Center for Intestinal and Liver Inflammation Research, Stanley Manne Children’s. Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - You-Yang Zhao
- grid.413808.60000 0004 0388 2248Program for Lung and Vascular Biology, Stanley Manne Children’s. Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL USA ,grid.16753.360000 0001 2299 3507Departments of Pediatrics, Pharmacology and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Xiao-Di Tan
- grid.16753.360000 0001 2299 3507Center for Intestinal and Liver Inflammation Research, Stanley Manne Children’s. Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL USA ,grid.16753.360000 0001 2299 3507Division of Gastroenterology, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Isabelle G. De Plaen
- grid.16753.360000 0001 2299 3507Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL USA ,grid.16753.360000 0001 2299 3507Center for Intestinal and Liver Inflammation Research, Stanley Manne Children’s. Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| |
Collapse
|
34
|
Hu S, Mahadevan A, Elysee IF, Choi J, Souchet NR, Bae GH, Taboada AK, Sanketi B, Duhamel GE, Sevier CS, Tao G, Kurpios NA. The asymmetric Pitx2 gene regulates gut muscular-lacteal development and protects against fatty liver disease. Cell Rep 2021; 37:110030. [PMID: 34818545 PMCID: PMC8650168 DOI: 10.1016/j.celrep.2021.110030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 08/19/2021] [Accepted: 10/29/2021] [Indexed: 12/25/2022] Open
Abstract
Intestinal lacteals are essential lymphatic channels for absorption and transport of dietary lipids and drive the pathogenesis of debilitating metabolic diseases. However, organ-specific mechanisms linking lymphatic dysfunction to disease etiology remain largely unknown. In this study, we uncover an intestinal lymphatic program that is linked to the left-right (LR) asymmetric transcription factor Pitx2. We show that deletion of the asymmetric Pitx2 enhancer ASE alters normal lacteal development through the lacteal-associated contractile smooth muscle lineage. ASE deletion leads to abnormal muscle morphogenesis induced by oxidative stress, resulting in impaired lacteal extension and defective lymphatic system-dependent lipid transport. Surprisingly, activation of lymphatic system-independent trafficking directs dietary lipids from the gut directly to the liver, causing diet-induced fatty liver disease. Our study reveals the molecular mechanism linking gut lymphatic function to the earliest symmetry-breaking Pitx2 and highlights the important relationship between intestinal lymphangiogenesis and the gut-liver axis.
Collapse
Affiliation(s)
- Shing Hu
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Aparna Mahadevan
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Isaac F Elysee
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Joseph Choi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Nathan R Souchet
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Gloria H Bae
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Alessandra K Taboada
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Bhargav Sanketi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Gerald E Duhamel
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Carolyn S Sevier
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA.
| |
Collapse
|
35
|
Almagro J, Messal HA, Zaw Thin M, van Rheenen J, Behrens A. Tissue clearing to examine tumour complexity in three dimensions. Nat Rev Cancer 2021; 21:718-730. [PMID: 34331034 DOI: 10.1038/s41568-021-00382-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
The visualization of whole organs and organisms through tissue clearing and fluorescence volumetric imaging has revolutionized the way we look at biological samples. Its application to solid tumours is changing our perception of tumour architecture, revealing signalling networks and cell interactions critical in tumour progression, and provides a powerful new strategy for cancer diagnostics. This Review introduces the latest advances in tissue clearing and three-dimensional imaging, examines the challenges in clearing epithelia - the tissue of origin of most malignancies - and discusses the insights that tissue clearing has brought to cancer research, as well as the prospective applications to experimental and clinical oncology.
Collapse
Affiliation(s)
- Jorge Almagro
- Adult Stem Cell Laboratory, The Francis Crick Institute, London, UK
| | - Hendrik A Messal
- Department of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - May Zaw Thin
- Cancer Stem Cell Laboratory, Institute of Cancer Research, London, UK
| | - Jacco van Rheenen
- Department of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Axel Behrens
- Adult Stem Cell Laboratory, The Francis Crick Institute, London, UK.
- Cancer Stem Cell Laboratory, Institute of Cancer Research, London, UK.
- Convergence Science Centre and Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK.
| |
Collapse
|
36
|
Huang Q, Garrett A, Bose S, Blocker S, Rios AC, Clevers H, Shen X. The frontier of live tissue imaging across space and time. Cell Stem Cell 2021; 28:603-622. [PMID: 33798422 PMCID: PMC8034393 DOI: 10.1016/j.stem.2021.02.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
What you see is what you get-imaging techniques have long been essential for visualization and understanding of tissue development, homeostasis, and regeneration, which are driven by stem cell self-renewal and differentiation. Advances in molecular and tissue modeling techniques in the last decade are providing new imaging modalities to explore tissue heterogeneity and plasticity. Here we describe current state-of-the-art imaging modalities for tissue research at multiple scales, with a focus on explaining key tradeoffs such as spatial resolution, penetration depth, capture time/frequency, and moieties. We explore emerging tissue modeling and molecular tools that improve resolution, specificity, and throughput.
Collapse
Affiliation(s)
- Qiang Huang
- Department of Pediatric Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004 Shaanxi, China; Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Aliesha Garrett
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Shree Bose
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Stephanie Blocker
- Center for In Vitro Microscopy, Duke University, Durham, NC 27708, USA
| | - Anne C Rios
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584, the Netherlands; Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht 3584, the Netherlands
| | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584, the Netherlands; Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht 3584, the Netherlands; Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, Utrecht 3584, the Netherlands
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
37
|
Sugimoto S, Kobayashi E, Fujii M, Ohta Y, Arai K, Matano M, Ishikawa K, Miyamoto K, Toshimitsu K, Takahashi S, Nanki K, Hakamata Y, Kanai T, Sato T. An organoid-based organ-repurposing approach to treat short bowel syndrome. Nature 2021; 592:99-104. [PMID: 33627870 DOI: 10.1038/s41586-021-03247-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 01/18/2021] [Indexed: 12/19/2022]
Abstract
The small intestine is the main organ for nutrient absorption, and its extensive resection leads to malabsorption and wasting conditions referred to as short bowel syndrome (SBS). Organoid technology enables an efficient expansion of intestinal epithelium tissue in vitro1, but reconstruction of the whole small intestine, including the complex lymphovascular system, has remained challenging2. Here we generate a functional small intestinalized colon (SIC) by replacing the native colonic epithelium with ileum-derived organoids. We first find that xenotransplanted human ileum organoids maintain their regional identity and form nascent villus structures in the mouse colon. In vitro culture of an organoid monolayer further reveals an essential role for luminal mechanistic flow in the formation of villi. We then develop a rat SIC model by repositioning the SIC at the ileocaecal junction, where the epithelium is exposed to a constant luminal stream of intestinal juice. This anatomical relocation provides the SIC with organ structures of the small intestine, including intact vasculature and innervation, villous structures, and the lacteal (a fat-absorbing lymphatic structure specific to the small intestine). The SIC has absorptive functions and markedly ameliorates intestinal failure in a rat model of SBS, whereas transplantation of colon organoids instead of ileum organoids invariably leads to mortality. These data provide a proof of principle for the use of intestinal organoids for regenerative purposes, and offer a feasible strategy for SBS treatment.
Collapse
Affiliation(s)
- Shinya Sugimoto
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan.,Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Eiji Kobayashi
- Department of Organ Fabrication, Keio University School of Medicine, Tokyo, Japan.
| | - Masayuki Fujii
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan.,Department of Surgical Oncology, The University of Tokyo, Tokyo, Japan
| | - Yuki Ohta
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kazuya Arai
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan.,JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Mami Matano
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Keiko Ishikawa
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan.,Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Kentaro Miyamoto
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan.,Miyarisan Pharmaceutical Co. Ltd, Tokyo, Japan
| | - Kohta Toshimitsu
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan.,Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Sirirat Takahashi
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kosaku Nanki
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan.,Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Yoji Hakamata
- School of Veterinary Nursing and Technology, Nippon Veterinary and Life Science University, Tokyo, Japan.,Research Center for Animal Life Science, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Takanori Kanai
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan. .,Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
38
|
Kim YS, Zhang H, Lee S, Park S, Noh M, Kim YM, Kwon YG. CU06-1004 Alleviates Experimental Colitis by Modulating Colonic Vessel Dysfunction. Front Pharmacol 2020; 11:571266. [PMID: 33041812 PMCID: PMC7523507 DOI: 10.3389/fphar.2020.571266] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
Inflammatory bowel disease is an autoimmune disease that causes chronic inflammation of the gastrointestinal tract. Endothelial dysfunction, defined by a reduced endothelial barrier and an increase in the expression of adhesion molecules, is part of the pathology of inflammatory bowel disease. In this study, we assessed the therapeutic effect of CU06-1004, an endothelial dysfunction blocker that reduces vascular hyperpermeability and inflammation in a mouse model of colitis. Acute colitis was induced in mice using 3% (w/v) dextran sodium sulfate added to their drinking water for 7 days. Twenty-four hours after the addition of dextran sodium sulfate, either mesalazine or CU06-1004 was administered orally each day. Administration of CU06-1004 significantly reduced the clinical manifestations (weight loss, diarrhea, and bloody stool) and histological changes (epithelium loss, inflammatory cell infiltration, and crypt destruction) induced by dextran sodium sulfate. Proinflammatory cytokines were also reduced, indicating that inflammation was ameliorated. From a vascular perspective, CU06-1004 reduced interrupted and tortuous vessels, enhanced junction protein expression, and reduced inflammatory adhesion molecules, indicating a broad improvement of endothelial dysfunction. Endothelial protection induced epithelial barrier restoration and decreased epithelial inflammation. Blocking endothelial dysfunction with CU06-1004 significantly ameliorated the progression of inflammatory bowel disease. Therefore, CU06-1004 may represent a potential therapeutic agent for the treatment of inflammatory bowel disease as well as other inflammatory diseases.
Collapse
Affiliation(s)
- Ye-Seul Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Haiying Zhang
- R&D Department, Curacle Co. Ltd, Seongnam-si, South Korea
| | - Sunghye Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Songyi Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Minyoung Noh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Young-Myeong Kim
- Vascular System Research Center, Kangwon National University, Chuncheon, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| |
Collapse
|
39
|
Duc D, Vigne S, Bernier-Latmani J, Yersin Y, Ruiz F, Gaïa N, Leo S, Lazarevic V, Schrenzel J, Petrova TV, Pot C. Disrupting Myelin-Specific Th17 Cell Gut Homing Confers Protection in an Adoptive Transfer Experimental Autoimmune Encephalomyelitis. Cell Rep 2020; 29:378-390.e4. [PMID: 31597098 DOI: 10.1016/j.celrep.2019.09.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 06/17/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is a common autoimmune disease of the CNS. Although an association between MS and inflammatory bowel diseases is observed, the link connecting intestinal immune responses and neuroinflammation remains unclear. Here we show that encephalitogenic Th17 cells infiltrate the colonic lamina propria before neurological symptom development in two murine MS models, active and adoptive transfer experimental autoimmune encephalomyelitis (EAE). Specifically targeting Th17 cell intestinal homing by blocking the α4β7-integrin and its ligand MAdCAM-1 pathway impairs T cell migration to the large intestine and dampens EAE severity in the Th17 cell adoptive transfer model. Mechanistically, myelin-specific Th17 cells proliferate in the colon and affect gut microbiota composition. The beneficial effect of blocking the α4β7-integrin and its ligand MAdCAM-1 pathway on EAE is interdependent with gut microbiota. Those results show that disrupting myelin-specific Th17 cell trafficking to the large intestine harnesses neuroinflammation and suggests that the gut environment and microbiota catalyze the encephalitogenic properties of Th17 cells.
Collapse
Affiliation(s)
- Donovan Duc
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Solenne Vigne
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Jeremiah Bernier-Latmani
- Department of Oncology, Lausanne University Hospital and University of Lausanne, and Ludwig Institute for Cancer Research Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Yannick Yersin
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Florian Ruiz
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Nadia Gaïa
- Genomic Research Laboratory, Division of Infectious Diseases, Department of Medicine, Geneva University Hospitals and University of Geneva, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva, Switzerland
| | - Stefano Leo
- Genomic Research Laboratory, Division of Infectious Diseases, Department of Medicine, Geneva University Hospitals and University of Geneva, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva, Switzerland
| | - Vladimir Lazarevic
- Genomic Research Laboratory, Division of Infectious Diseases, Department of Medicine, Geneva University Hospitals and University of Geneva, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva, Switzerland
| | - Jacques Schrenzel
- Genomic Research Laboratory, Division of Infectious Diseases, Department of Medicine, Geneva University Hospitals and University of Geneva, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, Lausanne University Hospital and University of Lausanne, and Ludwig Institute for Cancer Research Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Caroline Pot
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland.
| |
Collapse
|
40
|
McCarthy N, Kraiczy J, Shivdasani RA. Cellular and molecular architecture of the intestinal stem cell niche. Nat Cell Biol 2020; 22:1033-1041. [PMID: 32884148 DOI: 10.1038/s41556-020-0567-z] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/29/2020] [Indexed: 12/23/2022]
Abstract
Intestinal stem and progenitor cells replicate and differentiate in distinct compartments, influenced by Wnt, BMP, and other subepithelial cues. The cellular sources of these signals were long obscure because intestinal mesenchyme was insufficiently characterised. In this Review, we discuss how recent mRNA profiles of mouse and human intestinal submucosa, coupled with fine-resolution microscopy and gene and cell disruptions, reveal a coherent picture of an organised tissue carrying cells with distinct molecular properties and functions.
Collapse
Affiliation(s)
- Neil McCarthy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Judith Kraiczy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Petrova TV, Koh GY. Biological functions of lymphatic vessels. Science 2020; 369:369/6500/eaax4063. [PMID: 32646971 DOI: 10.1126/science.aax4063] [Citation(s) in RCA: 248] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
Abstract
The general functions of lymphatic vessels in fluid transport and immunosurveillance are well recognized. However, accumulating evidence indicates that lymphatic vessels play active and versatile roles in a tissue- and organ-specific manner during homeostasis and in multiple disease processes. This Review discusses recent advances to understand previously unidentified functions of adult mammalian lymphatic vessels, including immunosurveillance and immunomodulation upon pathogen invasion, transport of dietary fat, drainage of cerebrospinal fluid and aqueous humor, possible contributions toward neurodegenerative and neuroinflammatory diseases, and response to anticancer therapies.
Collapse
Affiliation(s)
- Tatiana V Petrova
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne and Centre Hospitalier Universitaire Vaudois, Chemin des Boveresses 155 CH-1066 Epalinges, Switzerland.
| | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science, Daejeon, 34141, Republic of Korea. .,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| |
Collapse
|
42
|
Krljanac B, Schubart C, Naumann R, Wirtz S, Culemann S, Krönke G, Voehringer D. RELMα-expressing macrophages protect against fatal lung damage and reduce parasite burden during helminth infection. Sci Immunol 2020; 4:4/35/eaau3814. [PMID: 31126996 DOI: 10.1126/sciimmunol.aau3814] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 01/08/2019] [Accepted: 04/26/2019] [Indexed: 12/15/2022]
Abstract
Alternatively activated macrophages (AAMs) can contribute to wound healing, regulation of glucose and fat metabolism, resolution of inflammation, and protective immunity against helminths. Their differentiation, tissue distribution, and effector functions are incompletely understood. Murine AAMs express high levels of resistin-like molecule (RELM) α, an effector protein with potent immunomodulatory functions. To visualize RELMα+ macrophages (MΦs) in vivo and evaluate their role in defense against helminths, we generated RELMα reporter/deleter mice. Infection with the helminth Nippostrongylus brasiliensis induced expansion of RELMα+ lung interstitial but not alveolar MΦs in a STAT6-dependent manner. RELMα+ MΦs were required for prevention of fatal lung damage during primary infection. Furthermore, protective immunity was lost upon specific deletion of RELMα+ MΦs during secondary infection. Thus, RELMα reporter/deleter mice reveal compartmentalization of AAMs in different tissues and demonstrate their critical role in resolution of severe lung inflammation and protection against migrating helminths.
Collapse
Affiliation(s)
- Branislav Krljanac
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Christoph Schubart
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Ronald Naumann
- Transgenic Core Facility, MPI of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Stefan Wirtz
- Department of Medicine 1-Gastroenterology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Stephan Culemann
- Department of Internal Medicine 3-Rheumatology and Immunology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3-Rheumatology and Immunology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany.
| |
Collapse
|
43
|
McCarthy N, Manieri E, Storm EE, Saadatpour A, Luoma AM, Kapoor VN, Madha S, Gaynor LT, Cox C, Keerthivasan S, Wucherpfennig K, Yuan GC, de Sauvage FJ, Turley SJ, Shivdasani RA. Distinct Mesenchymal Cell Populations Generate the Essential Intestinal BMP Signaling Gradient. Cell Stem Cell 2020; 26:391-402.e5. [PMID: 32084389 DOI: 10.1016/j.stem.2020.01.008] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/27/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022]
Abstract
Intestinal stem cells (ISCs) are confined to crypt bottoms and their progeny differentiate near crypt-villus junctions. Wnt and bone morphogenic protein (BMP) gradients drive this polarity, and colorectal cancer fundamentally reflects disruption of this homeostatic signaling. However, sub-epithelial sources of crucial agonists and antagonists that organize this BMP gradient remain obscure. Here, we couple whole-mount high-resolution microscopy with ensemble and single-cell RNA sequencing (RNA-seq) to identify three distinct PDGFRA+ mesenchymal cell types. PDGFRA(hi) telocytes are especially abundant at the villus base and provide a BMP reservoir, and we identified a CD81+ PDGFRA(lo) population present just below crypts that secretes the BMP antagonist Gremlin1. These cells, referred to as trophocytes, are sufficient to expand ISCs in vitro without additional trophic support and contribute to ISC maintenance in vivo. This study reveals intestinal mesenchymal structure at fine anatomic, molecular, and functional detail and the cellular basis for a signaling gradient necessary for tissue self-renewal.
Collapse
Affiliation(s)
- Neil McCarthy
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Elisa Manieri
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Elaine E Storm
- Department of Molecular Oncology, Genentech, South San Francisco, CA 94080, USA
| | - Assieh Saadatpour
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Adrienne M Luoma
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Varun N Kapoor
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Shariq Madha
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Liam T Gaynor
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Christian Cox
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Shilpa Keerthivasan
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Kai Wucherpfennig
- Department of Molecular Oncology, Genentech, South San Francisco, CA 94080, USA; Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Guo-Cheng Yuan
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | | | - Shannon J Turley
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA.
| |
Collapse
|
44
|
T Cell Recruitment to the Intestinal Stem Cell Compartment Drives Immune-Mediated Intestinal Damage after Allogeneic Transplantation. Immunity 2019; 51:90-103.e3. [PMID: 31278057 DOI: 10.1016/j.immuni.2019.06.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/01/2019] [Accepted: 06/05/2019] [Indexed: 12/30/2022]
Abstract
The key sites within the gastrointestinal (GI) tract where T cells mediate effector responses and the impact of these responses on intestinal stem cells (ISCs) remain unclear. Using experimental bone marrow transplantation to model immune-mediated GI damage and 3D imaging to analyze T cell localization, we found that the ISC compartment is the primary intestinal site targeted by T cells after transplantation. Recruitment to the crypt base region resulted in direct T cell engagement with the stem cell compartment and loss of crypt base columnar ISCs, which expressed both MHC classes I and II. Vasculature expressing the adhesion molecule MAdCAM-1 clustered near the crypt base, preferentially regulating crypt compartment invasion and ISC reduction without affecting T cell migration to villi. These findings indicate that allogeneic T cells rapidly access the stem cell niche after transplantation, and this targeted recruitment to the stem cell compartment results in ISC loss during immune-mediated GI damage.
Collapse
|
45
|
Abstract
The lacteal is a blunt-ended lymphatic capillary located at the center of a villus in the small intestine that plays multifaceted roles under both physiologic and pathologic conditions. However, studies of its biology are limited by the lack of a feasible method to visualize all the relevant components for its regulation. Here, we describe an efficient whole-mount protocol to visualize the intact structure of lacteals and surrounding cells in villi of the small intestine of adult mouse.
Collapse
|
46
|
Suh SH, Choe K, Hong SP, Jeong SH, Mäkinen T, Kim KS, Alitalo K, Surh CD, Koh GY, Song JH. Gut microbiota regulates lacteal integrity by inducing VEGF-C in intestinal villus macrophages. EMBO Rep 2019; 20:embr.201846927. [PMID: 30783017 DOI: 10.15252/embr.201846927] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/11/2019] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
A lacteal is a blunt-ended, long, tube-like lymphatic vessel located in the center of each intestinal villus that provides a unique route for drainage of absorbed lipids from the small intestine. However, key regulators for maintaining lacteal integrity are poorly understood. Here, we explore whether and how the gut microbiota regulates lacteal integrity. Germ depletion by antibiotic treatment triggers lacteal regression during adulthood and delays lacteal maturation during the postnatal period. In accordance with compromised lipid absorption, the button-like junction between lymphatic endothelial cells, which is ultrastructurally open to permit free entry of dietary lipids into lacteals, is significantly reduced in lacteals of germ-depleted mice. Lacteal defects are also found in germ-free mice, but conventionalization of germ-free mice leads to normalization of lacteals. Mechanistically, VEGF-C secreted from villus macrophages upon MyD88-dependent recognition of microbes and their products is a main factor in lacteal integrity. Collectively, we conclude that the gut microbiota is a crucial regulator for lacteal integrity by endowing its unique microenvironment and regulating villus macrophages in small intestine.
Collapse
Affiliation(s)
- Sang Heon Suh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Kibaek Choe
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Seon Pyo Hong
- Center for Vascular Research, Institute for Basic Science, Daejeon, Korea
| | - Seung-Hwan Jeong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kwang Soon Kim
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Korea
| | - Kari Alitalo
- Translational Cancer Biology Program and Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Charles D Surh
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, Korea
| | - Gou Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea .,Center for Vascular Research, Institute for Basic Science, Daejeon, Korea
| | - Joo-Hye Song
- Center for Vascular Research, Institute for Basic Science, Daejeon, Korea
| |
Collapse
|
47
|
Davis RB, Ding S, Nielsen NR, Pawlak JB, Blakeney ES, Caron KM. Calcitonin-Receptor-Like Receptor Signaling Governs Intestinal Lymphatic Innervation and Lipid Uptake. ACS Pharmacol Transl Sci 2019; 2:114-121. [PMID: 32219216 DOI: 10.1021/acsptsci.8b00061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Indexed: 02/06/2023]
Abstract
The absorption of dietary fat requires complex neuroendocrine-mediated regulation of chylomicron trafficking through enterocytes and intestinal lymphatic vessels. Calcitonin-receptor-like receptor (Calcrl) is a G protein-coupled receptor that can bind either a lymphangiogenic ligand adrenomedullin, with coreceptor RAMP2, or the neuropeptide CGRP, with coreceptor RAMP1. The extent to which this common GPCR controls lipid absorption via lymphatics or enteric innervation remains unclear. We used conditional and inducible genetic deletion of Calcrl in lymphatics to elucidate the pathophysiological consequences of this receptor pathway under conditions of high-fat diet. Inefficient absorption of dietary fat coupled with altered lymphatic endothelial junctions in Calcrl fl/fl /Prox1-CreER T2 mice results in excessive, transcellular lipid accumulation and abnormal enterocyte chylomicron processing and failure to gain weight. Interestingly, Calcrl fl/fl /Prox1-CreER T2 animals show reduced and disorganized mucosal and submucosal innervation. Consistently, mice with genetic loss of the CGRP coreceptor RAMP1 also displayed mucosal and submucosal innervation deficits, substantiating the CGRP-biased function of Calcrl in the neurolymphocrine axis. Thus, the common Calcrl receptor is a critical regulator of lipid absorption through its cell-specific functions in neurolymphocrine crosstalk.
Collapse
Affiliation(s)
- Reema B Davis
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| | - Shengli Ding
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| | - Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| | - John B Pawlak
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| | - Elizabeth S Blakeney
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| |
Collapse
|
48
|
Huang J, Brenna C, Khan AUM, Daniele C, Rudolf R, Heuveline V, Gretz N. A cationic near infrared fluorescent agent and ethyl-cinnamate tissue clearing protocol for vascular staining and imaging. Sci Rep 2019; 9:521. [PMID: 30679514 PMCID: PMC6345820 DOI: 10.1038/s41598-018-36741-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/23/2018] [Indexed: 12/20/2022] Open
Abstract
Understanding vascular structures and dysfunction is a fundamental challenge. This task has been approached by using traditional methodologies such as microscopic computed tomography and magnetic resonance imaging. Both techniques are not only expensive but also time-consuming. Here, we present a new method for visualizing vascular structures in different organs in an efficient manner. A cationic near infrared (NIR) fluorescent dye was developed with attractive features to specifically stain blood vessels. Furthermore, we refined the process of organ staining and harvesting by retrograde perfusion and optimized the subsequent dehydration and clearing process by the use of an automatic tissue processor and a non-toxic substance, ethyl-cinnamate. Using this approach, the time interval between organ harvesting and microscopic analysis can be reduced from day(s) or weeks to 4 hours. Finally, we have demonstrated that the new NIR fluorescent agent in combination with confocal or light-sheet microscopy can be efficiently used for visualization of vascular structures, such as the blood vessels in different organs e.g. glomeruli in kidneys, with an extremely high resolution. Our approach facilitates the development of automatic image processing and the quantitative analysis to study vascular and kidney diseases.
Collapse
Affiliation(s)
- Jiaguo Huang
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,Institute for Medical Technology, University of Heidelberg and University of Applied Sciences, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Cinzia Brenna
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,Institute for Medical Technology, University of Heidelberg and University of Applied Sciences, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Arif Ul Maula Khan
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,Institute for Medical Technology, University of Heidelberg and University of Applied Sciences, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Cristina Daniele
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,Institute for Medical Technology, University of Heidelberg and University of Applied Sciences, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Rüdiger Rudolf
- Institute for Medical Technology, University of Heidelberg and University of Applied Sciences, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163, Mannheim, Germany
| | - Vincent Heuveline
- Director of the Computing Centre, Heidelberg University, Im Neuenheimer Feld 293, 69120, Heidelberg, Germany
| | - Norbert Gretz
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany. .,Institute for Medical Technology, University of Heidelberg and University of Applied Sciences, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| |
Collapse
|
49
|
Cifarelli V, Eichmann A. The Intestinal Lymphatic System: Functions and Metabolic Implications. Cell Mol Gastroenterol Hepatol 2018; 7:503-513. [PMID: 30557701 PMCID: PMC6396433 DOI: 10.1016/j.jcmgh.2018.12.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 12/26/2022]
Abstract
The lymphatic system of the gut plays important roles in the transport of dietary lipids, as well as in immunosurveillance and removal of interstitial fluid. Historically, despite its crucial functions in intestinal homeostasis, the lymphatic system has been poorly studied. In the last 2 decades, identification of specific molecular mediators of lymphatic endothelial cells (LECs) growth together with novel genetic approaches and intravital imaging techniques, have advanced our understanding of the mechanisms regulating intestinal lymphatic physiology in health and disease. As its metabolic implications are gaining recognition, intestinal lymphatic biology is currently experiencing a surge in interest. This review describes current knowledge related to molecular control of intestinal lymphatic vessel structure and function. We discuss regulation of chylomicron entry into lymphatic vessels by vascular endothelial growth factors (VEGFs), hormones, transcription factors and the specific signaling pathways involved. The information covered supports the emerging role of intestinal lymphatics in etiology of the metabolic syndrome and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri,Correspondence Address correspondence to: Vincenza Cifarelli, PhD, Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, Campus box 8031, 660 Euclid Avenue, St. Louis, Missouri 63110. fax: (314) 362-8230.
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut,INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
50
|
Betterman KL, Harvey NL. Histological and Morphological Characterization of Developing Dermal Lymphatic Vessels. Methods Mol Biol 2018; 1846:19-35. [PMID: 30242750 DOI: 10.1007/978-1-4939-8712-2_2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The capacity to visualize the lymphatic vasculature in three-dimensions has revolutionized our understanding of the morphogenetic mechanisms important for constructing the lymphatic vascular network during development. Two complementary approaches are commonly employed to assess the function of genes and signaling pathways important for development of the dermal lymphatic vasculature in the mouse embryo. The first of these is whole-mount immunostaining of embryonic skin to analyze dermal lymphatic vessel network patterning and morphology in two and three dimensions. The second is immunostaining of thin tissue sections to examine lymphatic vessel identity, lumen formation and protein localization within discrete lymphatic endothelial cells in a two-dimensional setting. Here we present detailed protocols for multicolor immunofluorescent immunostaining of embryonic dorsal skin and thin tissue cryosections. Each of these methods generates high-resolution images of the dermal lymphatic vasculature, yielding information integral to in-depth characterization of lymphatic vessel phenotypes in the developing mouse embryo.
Collapse
Affiliation(s)
- Kelly L Betterman
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Natasha L Harvey
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.
| |
Collapse
|