1
|
Lins MP, de Melo IS. Exploring the interplay between cannabinoids and thymic functions. Toxicol Sci 2024; 202:1-12. [PMID: 39250730 DOI: 10.1093/toxsci/kfae114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024] Open
Abstract
Cannabinoids, derived from the Cannabis sativa plant, have garnered increasing attention for their potential therapeutic applications in various diseases. The pharmacologically active compounds in Cannabis, such as delta-9-tetrahydrocannabinol and cannabidiol, exhibit diverse immunomodulatory properties. Although studies have explored the effects of cannabinoids on immune function, their specific interactions with the thymus, a primary immune organ critical for T-cell development and maturation, remain an intriguing area of investigation. As the thymus plays a fundamental role in shaping the immune repertoire, understanding the interplay between cannabinoids and thymic function may shed light on potential benefits or concerns associated with Cannabis-based therapies. This article aims to provide an overview of the current scientific knowledge regarding the impact of medicinal Cannabis on the thymus and its implications for disease treatment and immune health.
Collapse
Affiliation(s)
- Marvin Paulo Lins
- Laboratory of Immunology, Department of Basic Sciences in Health, Faculty of Medicine, Federal University of Mato Grosso, Cuiabá-MT, 78060-900, Brazil
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, 21040-360, Brazil
| | - Igor Santana de Melo
- Laboratory of Neuropharmacology and Integrative Physiology, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, 57072-900, Brazil
| |
Collapse
|
2
|
Crosse EI, Binagui-Casas A, Gordon-Keylock S, Rybtsov S, Tamagno S, Olofsson D, Anderson RA, Medvinsky A. An interactive resource of molecular signalling in the developing human haematopoietic stem cell niche. Development 2023; 150:dev201972. [PMID: 37840454 PMCID: PMC10730088 DOI: 10.1242/dev.201972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023]
Abstract
The emergence of definitive human haematopoietic stem cells (HSCs) from Carnegie Stage (CS) 14 to CS17 in the aorta-gonad-mesonephros (AGM) region is a tightly regulated process. Previously, we conducted spatial transcriptomic analysis of the human AGM region at the end of this period (CS16/CS17) and identified secreted factors involved in HSC development. Here, we extend our analysis to investigate the progression of dorso-ventral polarised signalling around the dorsal aorta over the entire period of HSC emergence. Our results reveal a dramatic increase in ventral signalling complexity from the CS13-CS14 transition, coinciding with the first appearance of definitive HSCs. We further observe stage-specific changes in signalling up to CS17, which may underpin the step-wise maturation of HSCs described in the mouse model. The data-rich resource is also presented in an online interface enabling in silico analysis of molecular interactions between spatially defined domains of the AGM region. This resource will be of particular interest for researchers studying mechanisms underlying human HSC development as well as those developing in vitro methods for the generation of clinically relevant HSCs from pluripotent stem cells.
Collapse
Affiliation(s)
- Edie I. Crosse
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Anahi Binagui-Casas
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | | | - Stanislav Rybtsov
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Sara Tamagno
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Didrik Olofsson
- Omiqa Bioinformatics GmbH, Altensteinstraße 40, 14195 Berlin, Germany
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Alexander Medvinsky
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
3
|
Kouchaeknejad A, Van Der Walt G, De Donato MH, Puighermanal E. Imaging and Genetic Tools for the Investigation of the Endocannabinoid System in the CNS. Int J Mol Sci 2023; 24:15829. [PMID: 37958825 PMCID: PMC10648052 DOI: 10.3390/ijms242115829] [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: 09/15/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
As central nervous system (CNS)-related disorders present an increasing cause of global morbidity, mortality, and high pressure on our healthcare system, there is an urgent need for new insights and treatment options. The endocannabinoid system (ECS) is a critical network of endogenous compounds, receptors, and enzymes that contribute to CNS development and regulation. Given its multifaceted involvement in neurobiology and its significance in various CNS disorders, the ECS as a whole is considered a promising therapeutic target. Despite significant advances in our understanding of the ECS's role in the CNS, its complex architecture and extensive crosstalk with other biological systems present challenges for research and clinical advancements. To bridge these knowledge gaps and unlock the full therapeutic potential of ECS interventions in CNS-related disorders, a plethora of molecular-genetic tools have been developed in recent years. Here, we review some of the most impactful tools for investigating the neurological aspects of the ECS. We first provide a brief introduction to the ECS components, including cannabinoid receptors, endocannabinoids, and metabolic enzymes, emphasizing their complexity. This is followed by an exploration of cutting-edge imaging tools and genetic models aimed at elucidating the roles of these principal ECS components. Special emphasis is placed on their relevance in the context of CNS and its associated disorders.
Collapse
Affiliation(s)
| | | | | | - Emma Puighermanal
- Neuroscience Institute, Autonomous University of Barcelona, 08193 Bellaterra, Spain; (A.K.); (G.V.D.W.); (M.H.D.D.)
| |
Collapse
|
4
|
Ishida T, Heck AM, Varnum-Finney B, Dozono S, Nourigat-McKay C, Kraskouskas K, Wellington R, Waltner O, Root, Jackson DL, Delaney C, Rafii S, Bernstein ID, Trapnell, Hadland B. Differentiation latency and dormancy signatures define fetal liver HSCs at single cell resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.01.543314. [PMID: 37333272 PMCID: PMC10274697 DOI: 10.1101/2023.06.01.543314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Decoding the gene regulatory mechanisms mediating self-renewal of hematopoietic stem cells (HSCs) during their amplification in the fetal liver (FL) is relevant for advancing therapeutic applications aiming to expand transplantable HSCs, a long-standing challenge. Here, to explore intrinsic and extrinsic regulation of self-renewal in FL-HSCs at the single cell level, we engineered a culture platform designed to recapitulate the FL endothelial niche, which supports the amplification of serially engraftable HSCs ex vivo. Leveraging this platform in combination with single cell index flow cytometry, serial transplantation assays, and single cell RNA-sequencing, we elucidated previously unrecognized heterogeneity in immunophenotypically defined FL-HSCs and demonstrated that differentiation latency and transcriptional signatures of biosynthetic dormancy are distinguishing properties of self-renewing FL-HSCs with capacity for serial, long-term multilineage hematopoietic reconstitution. Altogether, our findings provide key insights into HSC expansion and generate a novel resource for future exploration of the intrinsic and niche-derived signaling pathways that support FL-HSC self-renewal.
Collapse
Affiliation(s)
- Takashi Ishida
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Adam M. Heck
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Barbara Varnum-Finney
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Stacey Dozono
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Cynthia Nourigat-McKay
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Katie Kraskouskas
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Rachel Wellington
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology, School of Medicine, University of Washington, Seattle, WA
| | - Olivia Waltner
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Root
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dana L Jackson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Colleen Delaney
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Deverra Therapeutics, Seattle, WA, USA
- Division of Pediatric Hematology/Oncology, University of Washington, Seattle, WA, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Irwin D. Bernstein
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Pediatric Hematology/Oncology, University of Washington, Seattle, WA, USA
| | - Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Brandon Hadland
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Pediatric Hematology/Oncology, University of Washington, Seattle, WA, USA
| |
Collapse
|
5
|
Arceri L, Nguyen TK, Gibson S, Baker S, Wingert RA. Cannabinoid Signaling in Kidney Disease. Cells 2023; 12:1419. [PMID: 37408253 DOI: 10.3390/cells12101419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 07/07/2023] Open
Abstract
Endocannabinoid signaling plays crucial roles in human physiology in the function of multiple systems. The two cannabinoid receptors, CB1 and CB2, are cell membrane proteins that interact with both exogenous and endogenous bioactive lipid ligands, or endocannabinoids. Recent evidence has established that endocannabinoid signaling operates within the human kidney, as well as suggests the important role it plays in multiple renal pathologies. CB1, specifically, has been identified as the more prominent ECS receptor within the kidney, allowing us to place emphasis on this receptor. The activity of CB1 has been repeatedly shown to contribute to both diabetic and non-diabetic chronic kidney disease (CKD). Interestingly, recent reports of acute kidney injury (AKI) have been attributed to synthetic cannabinoid use. Therefore, the exploration of the ECS, its receptors, and its ligands can help provide better insight into new methods of treatment for a range of renal diseases. This review explores the endocannabinoid system, with a focus on its impacts within the healthy and diseased kidney.
Collapse
Affiliation(s)
- Liana Arceri
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Thanh Khoa Nguyen
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Shannon Gibson
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Sophia Baker
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
6
|
Ding J, Li Y, Larochelle A. De Novo Generation of Human Hematopoietic Stem Cells from Pluripotent Stem Cells for Cellular Therapy. Cells 2023; 12:321. [PMID: 36672255 PMCID: PMC9857267 DOI: 10.3390/cells12020321] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
The ability to manufacture human hematopoietic stem cells (HSCs) in the laboratory holds enormous promise for cellular therapy of human blood diseases. Several differentiation protocols have been developed to facilitate the emergence of HSCs from human pluripotent stem cells (PSCs). Most approaches employ a stepwise addition of cytokines and morphogens to recapitulate the natural developmental process. However, these protocols globally lack clinical relevance and uniformly induce PSCs to produce hematopoietic progenitors with embryonic features and limited engraftment and differentiation capabilities. This review examines how key intrinsic cues and extrinsic environmental inputs have been integrated within human PSC differentiation protocols to enhance the emergence of definitive hematopoiesis and how advances in genomics set the stage for imminent breakthroughs in this field.
Collapse
Affiliation(s)
| | | | - Andre Larochelle
- Cellular and Molecular Therapeutics Branch, National Heart Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Cacialli P, Mailhe MP, Wagner I, Merkler D, Golub R, Bertrand JY. Synergistic prostaglandin E synthesis by myeloid and endothelial cells promotes fetal hematopoietic stem cell expansion in vertebrates. EMBO J 2022; 41:e108536. [PMID: 35924455 PMCID: PMC9531293 DOI: 10.15252/embj.2021108536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/21/2022] Open
Abstract
During development, hematopoietic stem cells (HSCs) are produced from the hemogenic endothelium and will expand in a transient hematopoietic niche. Prostaglandin E2 (PGE2) is essential during vertebrate development and HSC specification, but its precise source in the embryo remains elusive. Here, we show that in the zebrafish embryo, PGE2 synthesis genes are expressed by distinct stromal cell populations, myeloid (neutrophils, macrophages), and endothelial cells of the caudal hematopoietic tissue. Ablation of myeloid cells, which produce the PGE2 precursor prostaglandin H2 (PGH2), results in loss of HSCs in the caudal hematopoietic tissue, which could be rescued by exogeneous PGE2 or PGH2 supplementation. Endothelial cells contribute by expressing the PGH2 import transporter slco2b1 and ptges3, the enzyme converting PGH2 into PGE2. Of note, differential niche cell expression of PGE2 biosynthesis enzymes is also observed in the mouse fetal liver. Taken altogether, our data suggest that the triad composed of neutrophils, macrophages, and endothelial cells sequentially and synergistically contributes to blood stem cell expansion during vertebrate development.
Collapse
Affiliation(s)
- Pietro Cacialli
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva 4, Switzerland
| | | | - Ingrid Wagner
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva 4, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva 4, Switzerland.,Division of Clinical Pathology, Department of Diagnostic, University Hospitals of Geneva, Geneva, Switzerland
| | - Rachel Golub
- Unité Lymphocytes et Immunité, Pasteur Institute, Paris Cedex 15, France.,Université de Paris, Paris, France
| | - Julien Y Bertrand
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva 4, Switzerland
| |
Collapse
|
8
|
Gazzi T, Brennecke B, Atz K, Korn C, Sykes D, Forn-Cuni G, Pfaff P, Sarott RC, Westphal MV, Mostinski Y, Mach L, Wasinska-Kalwa M, Weise M, Hoare BL, Miljuš T, Mexi M, Roth N, Koers EJ, Guba W, Alker A, Rufer AC, Kusznir EA, Huber S, Raposo C, Zirwes EA, Osterwald A, Pavlovic A, Moes S, Beck J, Nettekoven M, Benito-Cuesta I, Grande T, Drawnel F, Widmer G, Holzer D, van der Wel T, Mandhair H, Honer M, Fingerle J, Scheffel J, Broichhagen J, Gawrisch K, Romero J, Hillard CJ, Varga ZV, van der Stelt M, Pacher P, Gertsch J, Ullmer C, McCormick PJ, Oddi S, Spaink HP, Maccarrone M, Veprintsev DB, Carreira EM, Grether U, Nazaré M. Detection of cannabinoid receptor type 2 in native cells and zebrafish with a highly potent, cell-permeable fluorescent probe. Chem Sci 2022; 13:5539-5545. [PMID: 35694350 PMCID: PMC9116301 DOI: 10.1039/d1sc06659e] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/22/2022] [Indexed: 12/16/2022] Open
Abstract
Despite its essential role in the (patho)physiology of several diseases, CB2R tissue expression profiles and signaling mechanisms are not yet fully understood. We report the development of a highly potent, fluorescent CB2R agonist probe employing structure-based reverse design. It commences with a highly potent, preclinically validated ligand, which is conjugated to a silicon-rhodamine fluorophore, enabling cell permeability. The probe is the first to preserve interspecies affinity and selectivity for both mouse and human CB2R. Extensive cross-validation (FACS, TR-FRET and confocal microscopy) set the stage for CB2R detection in endogenously expressing living cells along with zebrafish larvae. Together, these findings will benefit clinical translatability of CB2R based drugs.
Collapse
Affiliation(s)
- Thais Gazzi
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Benjamin Brennecke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Kenneth Atz
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Claudia Korn
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - David Sykes
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK
- United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | | | - Patrick Pfaff
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich Vladimir-Prelog-Weg 3 8093 Zürich Switzerland
| | - Roman C Sarott
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich Vladimir-Prelog-Weg 3 8093 Zürich Switzerland
| | - Matthias V Westphal
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich Vladimir-Prelog-Weg 3 8093 Zürich Switzerland
| | - Yelena Mostinski
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Leonard Mach
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Malgorzata Wasinska-Kalwa
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Marie Weise
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Bradley L Hoare
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK
- United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | - Tamara Miljuš
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK
- United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | - Maira Mexi
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK
- United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | - Nicolas Roth
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London London EC1M 6BQ England UK
| | - Eline J Koers
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK
- United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | - Wolfgang Guba
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - André Alker
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Arne C Rufer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Eric A Kusznir
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Sylwia Huber
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Catarina Raposo
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Elisabeth A Zirwes
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Anja Osterwald
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Anto Pavlovic
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Svenja Moes
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Jennifer Beck
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Matthias Nettekoven
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Irene Benito-Cuesta
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria Pozuelo de Alarcón 28223 Madrid Spain
| | - Teresa Grande
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria Pozuelo de Alarcón 28223 Madrid Spain
| | - Faye Drawnel
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Gabriella Widmer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Daniela Holzer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Tom van der Wel
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University 2333 CC Leiden the Netherlands
| | - Harpreet Mandhair
- Institute of Biochemistry and Molecular Medicine, University of Bern 3012 Bern Switzerland
| | - Michael Honer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Jürgen Fingerle
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Jörg Scheffel
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin Berlin Germany
- Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP Berlin Germany
| | - Johannes Broichhagen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Klaus Gawrisch
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health Rockville MD 20852 USA
| | - Julián Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria Pozuelo de Alarcón 28223 Madrid Spain
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology, Neuroscience Research Center, Medical College of Wisconsin Milwaukee WI 53226 USA
| | - Zoltan V Varga
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health Rockville MD 20852 USA
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University 1085 Budapest Hungary
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University 2333 CC Leiden the Netherlands
| | - Pal Pacher
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health Rockville MD 20852 USA
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, University of Bern 3012 Bern Switzerland
| | - Christoph Ullmer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Peter J McCormick
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London London EC1M 6BQ England UK
| | - Sergio Oddi
- Faculty of Veterinary Medicine, University of Teramo 64100 Teramo European Italy
- European Center for Brain Research (CERC), Santa Lucia Foundation 00179 Rome Italy
| | - Herman P Spaink
- Leiden University Einsteinweg 55 2333 CC Leiden the Netherlands
| | - Mauro Maccarrone
- European Center for Brain Research (CERC), Santa Lucia Foundation 00179 Rome Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila 67100 L'Aquila Italy
| | - Dmitry B Veprintsev
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK
- United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | - Erick M Carreira
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich Vladimir-Prelog-Weg 3 8093 Zürich Switzerland
| | - Uwe Grether
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Marc Nazaré
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| |
Collapse
|
9
|
Hadland B, Varnum-Finney B, Dozono S, Dignum T, Nourigat-McKay C, Heck AM, Ishida T, Jackson DL, Itkin T, Butler JM, Rafii S, Trapnell C, Bernstein ID. Engineering a niche supporting hematopoietic stem cell development using integrated single-cell transcriptomics. Nat Commun 2022; 13:1584. [PMID: 35332125 PMCID: PMC8948249 DOI: 10.1038/s41467-022-28781-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 02/09/2022] [Indexed: 12/22/2022] Open
Abstract
Hematopoietic stem cells (HSCs) develop from hemogenic endothelium within embryonic arterial vessels such as the aorta of the aorta-gonad-mesonephros region (AGM). To identify the signals responsible for HSC formation, here we use single cell RNA-sequencing to simultaneously analyze the transcriptional profiles of AGM-derived cells transitioning from hemogenic endothelium to HSCs, and AGM-derived endothelial cells which provide signals sufficient to support HSC maturation and self-renewal. Pseudotemporal ordering reveals dynamics of gene expression during the hemogenic endothelium to HSC transition, identifying surface receptors specifically expressed on developing HSCs. Transcriptional profiling of niche endothelial cells identifies corresponding ligands, including those signaling to Notch receptors, VLA-4 integrin, and CXCR4, which, when integrated in an engineered platform, are sufficient to support the generation of engrafting HSCs. These studies provide a transcriptional map of the signaling interactions necessary for the development of HSCs and advance the goal of engineering HSCs for therapeutic applications.
Collapse
Affiliation(s)
- Brandon Hadland
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98105, USA.
| | - Barbara Varnum-Finney
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Stacey Dozono
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Tessa Dignum
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Cynthia Nourigat-McKay
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Adam M Heck
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Takashi Ishida
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Dana L Jackson
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, 98105, USA
| | - Tomer Itkin
- Department of Genetic Medicine, Ansary Stem Cell Institute, Howard Hughes Medical Institute, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Jason M Butler
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ, 07110, USA
| | - Shahin Rafii
- Department of Genetic Medicine, Ansary Stem Cell Institute, Howard Hughes Medical Institute, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, 98105, USA
| | - Irwin D Bernstein
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98105, USA
| |
Collapse
|
10
|
Park YM, Dahlem C, Meyer MR, Kiemer AK, Müller R, Herrmann J. Induction of Liver Size Reduction in Zebrafish Larvae by the Emerging Synthetic Cannabinoid 4F-MDMB-BINACA and Its Impact on Drug Metabolism. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041290. [PMID: 35209079 PMCID: PMC8879502 DOI: 10.3390/molecules27041290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/22/2022]
Abstract
Zebrafish (ZF; Danio rerio) larvae have become a popular in vivo model in drug metabolism studies. Here, we investigated the metabolism of methyl 2-[1-(4-fluorobutyl)-1H-indazole-3-carboxamido]-3,3-dimethylbutanoate (4F-MDMB-BINACA) in ZF larvae after direct administration of the cannabinoid via microinjection, and we visualized the spatial distributions of the parent compound and its metabolites by mass spectrometry imaging (MSI). Furthermore, using genetically modified ZF larvae, the role of cannabinoid receptor type 1 (CB1) and type 2 (CB2) on drug metabolism was studied. Receptor-deficient ZF mutant larvae were created using morpholino oligonucleotides (MOs), and CB2-deficiency had a critical impact on liver development of ZF larva, leading to a significant reduction of liver size. A similar phenotype was observed when treating wild-type ZF larvae with 4F-MDMB-BINACA. Thus, we reasoned that the cannabinoid-induced impaired liver development might also influence its metabolic function. Studying the metabolism of two synthetic cannabinoids, 4F-MDMB-BINACA and methyl 2-(1-(5-fluoropentyl)-1H-pyrrolo[2,3-b]pyridine-3-carboxamido)-3,3-dimethylbutanoate (7′N-5F-ADB), revealed important insights into the in vivo metabolism of these compounds and the role of cannabinoid receptor binding.
Collapse
Affiliation(s)
- Yu Mi Park
- Helmholtz Centre for Infection Research, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8 1, Saarland University, 66123 Saarbrücken, Germany;
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Charlotte Dahlem
- Department of Pharmacy, Pharmaceutical Biology, Campus C2 3, Saarland University, 66123 Saarbrücken, Germany; (C.D.); (A.K.K.)
| | - Markus R. Meyer
- Center for Molecular Signaling (PZMS), Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Experimental and Clinical Toxicology, Saarland University, 66421 Homburg, Germany;
| | - Alexandra K. Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Campus C2 3, Saarland University, 66123 Saarbrücken, Germany; (C.D.); (A.K.K.)
| | - Rolf Müller
- Helmholtz Centre for Infection Research, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8 1, Saarland University, 66123 Saarbrücken, Germany;
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
- German Center for Infection Research (DZIF), 38124 Braunschweig, Germany
- Correspondence: (R.M.); (J.H.)
| | - Jennifer Herrmann
- Helmholtz Centre for Infection Research, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8 1, Saarland University, 66123 Saarbrücken, Germany;
- German Center for Infection Research (DZIF), 38124 Braunschweig, Germany
- Correspondence: (R.M.); (J.H.)
| |
Collapse
|
11
|
Sugden WW, North TE. Making Blood from the Vessel: Extrinsic and Environmental Cues Guiding the Endothelial-to-Hematopoietic Transition. Life (Basel) 2021; 11:life11101027. [PMID: 34685398 PMCID: PMC8539454 DOI: 10.3390/life11101027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 01/10/2023] Open
Abstract
It is increasingly recognized that specialized subsets of endothelial cells carry out unique functions in specific organs and regions of the vascular tree. Perhaps the most striking example of this specialization is the ability to contribute to the generation of the blood system, in which a distinct population of “hemogenic” endothelial cells in the embryo transforms irreversibly into hematopoietic stem and progenitor cells that produce circulating erythroid, myeloid and lymphoid cells for the lifetime of an animal. This review will focus on recent advances made in the zebrafish model organism uncovering the extrinsic and environmental factors that facilitate hemogenic commitment and the process of endothelial-to-hematopoietic transition that produces blood stem cells. We highlight in particular biomechanical influences of hemodynamic forces and the extracellular matrix, metabolic and sterile inflammatory cues present during this developmental stage, and outline new avenues opened by transcriptomic-based approaches to decipher cell–cell communication mechanisms as examples of key signals in the embryonic niche that regulate hematopoiesis.
Collapse
Affiliation(s)
- Wade W. Sugden
- Stem Cell Program, Department of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA;
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
| | - Trista E. North
- Stem Cell Program, Department of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA;
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| |
Collapse
|
12
|
Chambers JM, Addiego A, Flores-Mireles AL, Wingert RA. Ppargc1a Controls Ciliated Cell Development by Regulating Prostaglandin Biosynthesis. Cell Rep 2020; 33:108370. [PMID: 33176142 PMCID: PMC7731726 DOI: 10.1016/j.celrep.2020.108370] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 07/23/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022] Open
Abstract
Cilia are microtubule-based organelles that function in a multitude of physiological contexts to perform chemosensing, mechanosensing, and fluid propulsion. The process of ciliogenesis is highly regulated, and disruptions result in disease states termed ciliopathies. Here, we report that peroxisome proliferator-activated receptor gamma, coactivator 1 alpha (ppargc1a) is essential for ciliogenesis in nodal, mono-, and multiciliated cells (MCCs) and for discernment of renal tubule ciliated cell fate during embryogenesis. ppargc1a performs these functions by affecting prostaglandin signaling, whereby cilia formation and renal MCC fate are restored with prostaglandin E2 (PGE2) treatment in ppargc1a-deficient animals. Genetic disruption of ppargc1a specifically reduces expression of the prostanoid biosynthesis gene prostaglandin-endoperoxide synthase 1 (ptgs1), and suboptimal knockdown of both genes shows this synergistic effect. Furthermore, ptgs1 overexpression rescues ciliogenesis and renal MCCs in ppargc1a-deficient embryos. These findings position Ppargc1a as a key genetic regulator of prostaglandin signaling during ciliated cell ontogeny.
Collapse
Affiliation(s)
- Joseph M Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Amanda Addiego
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Ana L Flores-Mireles
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
13
|
Frame JM, Kubaczka C, Long TL, Esain V, Soto RA, Hachimi M, Jing R, Shwartz A, Goessling W, Daley GQ, North TE. Metabolic Regulation of Inflammasome Activity Controls Embryonic Hematopoietic Stem and Progenitor Cell Production. Dev Cell 2020; 55:133-149.e6. [PMID: 32810442 DOI: 10.1016/j.devcel.2020.07.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 05/26/2020] [Accepted: 07/22/2020] [Indexed: 12/21/2022]
Abstract
Embryonic hematopoietic stem and progenitor cells (HSPCs) robustly proliferate while maintaining multilineage potential in vivo; however, an incomplete understanding of spatiotemporal cues governing their generation has impeded robust production from human induced pluripotent stem cells (iPSCs) in vitro. Using the zebrafish model, we demonstrate that NLRP3 inflammasome-mediated interleukin-1-beta (IL1β) signaling drives HSPC production in response to metabolic activity. Genetic induction of active IL1β or pharmacologic inflammasome stimulation increased HSPC number as assessed by in situ hybridization for runx1/cmyb and flow cytometry. Loss of inflammasome components, including il1b, reduced CD41+ HSPCs and prevented their expansion in response to metabolic cues. Cell ablation studies indicated that macrophages were essential for initial inflammasome stimulation of Il1rl1+ HSPCs. Significantly, in human iPSC-derived hemogenic precursors, transient inflammasome stimulation increased multilineage hematopoietic colony-forming units and T cell progenitors. This work establishes the inflammasome as a conserved metabolic sensor that expands HSPC production in vivo and in vitro.
Collapse
Affiliation(s)
- Jenna M Frame
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Caroline Kubaczka
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Timothy L Long
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Virginie Esain
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Rebecca A Soto
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
| | - Mariam Hachimi
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Ran Jing
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Arkadi Shwartz
- Genetics Division, Brigham & Women's Hospital, Boston, MA 02115, USA
| | - Wolfram Goessling
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA; Genetics Division, Brigham & Women's Hospital, Boston, MA 02115, USA; Gastroenterology Division, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - George Q Daley
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Trista E North
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
14
|
Marijuana and Opioid Use during Pregnancy: Using Zebrafish to Gain Understanding of Congenital Anomalies Caused by Drug Exposure during Development. Biomedicines 2020; 8:biomedicines8080279. [PMID: 32784457 PMCID: PMC7460517 DOI: 10.3390/biomedicines8080279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 01/09/2023] Open
Abstract
Marijuana and opioid addictions have increased alarmingly in recent decades, especially in the United States, posing threats to society. When the drug user is a pregnant mother, there is a serious risk to the developing baby. Congenital anomalies are associated with prenatal exposure to marijuana and opioids. Here, we summarize the current data on the prevalence of marijuana and opioid use among the people of the United States, particularly pregnant mothers. We also summarize the current zebrafish studies used to model and understand the effects of these drug exposures during development and to understand the behavioral changes after exposure. Zebrafish experiments recapitulate the drug effects seen in human addicts and the birth defects seen in human babies prenatally exposed to marijuana and opioids. Zebrafish show great potential as an easy and inexpensive model for screening compounds for their ability to mitigate the drug effects, which could lead to new therapeutics.
Collapse
|
15
|
Rose CD, Pompili D, Henke K, Van Gennip JLM, Meyer-Miner A, Rana R, Gobron S, Harris MP, Nitz M, Ciruna B. SCO-Spondin Defects and Neuroinflammation Are Conserved Mechanisms Driving Spinal Deformity across Genetic Models of Idiopathic Scoliosis. Curr Biol 2020; 30:2363-2373.e6. [PMID: 32386528 DOI: 10.1016/j.cub.2020.04.020] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/05/2020] [Accepted: 04/08/2020] [Indexed: 12/23/2022]
Abstract
Adolescent idiopathic scoliosis (AIS) affects 3% to 4% of children between the ages of 11 and 18 [1, 2]. This disorder, characterized by abnormal three-dimensional spinal curvatures that typically develop during periods of rapid growth, occurs in the absence of congenital vertebral malformations or neuromuscular defects [1]. Genetic heterogeneity [3] and a historical lack of appropriate animal models [4] have confounded basic understanding of AIS biology; thus, treatment options remain limited [5, 6]. Recently, genetic studies using zebrafish have linked idiopathic-like scoliosis to irregularities in motile cilia-mediated cerebrospinal fluid flow [7-9]. However, because loss of cilia motility in human primary ciliary dyskinesia patients is not fully associated with scoliosis [10, 11], other pathogenic mechanisms remain to be determined. Here, we demonstrate that zebrafish scospondin (sspo) mutants develop late-onset idiopathic-like spinal curvatures in the absence of obvious cilia motility defects. Sspo is a large secreted glycoprotein functionally associated with the subcommissural organ and Reissner's fiber [12]-ancient and enigmatic organs of the brain ventricular system reported to govern cerebrospinal fluid homeostasis [13, 14], neurogenesis [12, 15-18], and embryonic morphogenesis [19]. We demonstrate that irregular deposition of Sspo within brain ventricles is associated with idiopathic-like scoliosis across diverse genetic models. Furthermore, Sspo defects are sufficient to induce oxidative stress and neuroinflammatory responses implicated in AIS pathogenesis [9]. Through screening for chemical suppressors of sspo mutant phenotypes, we also identify potent agents capable of blocking severe juvenile spine deformity. Our work thus defines a new preclinical model of AIS and provides tools to realize novel therapeutic strategies.
Collapse
Affiliation(s)
- Chloe D Rose
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David Pompili
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Katrin Henke
- Department of Orthopedic Research, Boston Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Jenica L M Van Gennip
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Anne Meyer-Miner
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Rahul Rana
- Department of Chemistry, The University of Toronto, Toronto, ON M5S 3H6, Canada
| | | | - Matthew P Harris
- Department of Orthopedic Research, Boston Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Mark Nitz
- Department of Chemistry, The University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Brian Ciruna
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, The University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
16
|
Lu X, Zhang Y, Liu F, Wang L. Rac2 Regulates the Migration of T Lymphoid Progenitors to the Thymus during Zebrafish Embryogenesis. THE JOURNAL OF IMMUNOLOGY 2020; 204:2447-2454. [PMID: 32198141 DOI: 10.4049/jimmunol.1901494] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/12/2020] [Indexed: 12/30/2022]
Abstract
The caudal hematopoietic tissue in zebrafish, the equivalent to the fetal liver in mammals, is an intermediate hematopoietic niche for the maintenance and differentiation of hematopoietic stem and progenitor cells before homing to the thymus and kidney marrow. As one of the ultimate hematopoietic organs, the thymus sustains T lymphopoiesis, which is essential for adaptive immune system. However, the mechanism of prethymic T lymphoid progenitors migrating to the thymus remains elusive. In this study, we identify an Rho GTPase Rac2 as a modulator of T lymphoid progenitor homing to the thymus in zebrafish. rac2-Deficient embryos show the inability of T lymphoid progenitors homing to the thymus because of defective cell-autonomous motility. Mechanistically, we demonstrate that Rac2 regulates homing of T lymphoid progenitor through Pak1-mediated AKT pathway. Taken together, our work reveals an important function of Rac2 in directing T lymphoid progenitor migration to the thymus during zebrafish embryogenesis.
Collapse
Affiliation(s)
- Xinyan Lu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; and.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanlin Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; and
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; and .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lu Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China;
| |
Collapse
|
17
|
Lundin V, Sugden WW, Theodore LN, Sousa PM, Han A, Chou S, Wrighton PJ, Cox AG, Ingber DE, Goessling W, Daley GQ, North TE. YAP Regulates Hematopoietic Stem Cell Formation in Response to the Biomechanical Forces of Blood Flow. Dev Cell 2020; 52:446-460.e5. [PMID: 32032546 DOI: 10.1016/j.devcel.2020.01.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 10/16/2019] [Accepted: 01/07/2020] [Indexed: 12/27/2022]
Abstract
Hematopoietic stem and progenitor cells (HSPCs), first specified from hemogenic endothelium (HE) in the ventral dorsal aorta (VDA), support lifelong hematopoiesis. Their de novo production promises significant therapeutic value; however, current in vitro approaches cannot efficiently generate multipotent long-lived HSPCs. Presuming this reflects a lack of extrinsic cues normally impacting the VDA, we devised a human dorsal aorta-on-a-chip platform that identified Yes-activated protein (YAP) as a cyclic stretch-induced regulator of HSPC formation. In the zebrafish VDA, inducible Yap overexpression significantly increased runx1 expression in vivo and the number of CD41+ HSPCs downstream of HE specification. Endogenous Yap activation by lats1/2 knockdown or Rho-GTPase stimulation mimicked Yap overexpression and induced HSPCs in embryos lacking blood flow. Notably, in static human induced pluripotent stem cell (iPSC)-derived HE culture, compound-mediated YAP activation enhanced RUNX1 levels and hematopoietic colony-forming potential. Together, our findings reveal a potent impact of hemodynamic Rho-YAP mechanotransduction on HE fate, relevant to de novo human HSPC production.
Collapse
Affiliation(s)
- Vanessa Lundin
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Wade W Sugden
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Lindsay N Theodore
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Patricia M Sousa
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Areum Han
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Stephanie Chou
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Paul J Wrighton
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew G Cox
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Wolfram Goessling
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA 02115, USA
| | - George Q Daley
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Trista E North
- Stem Cell Program, Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
18
|
Sufian MS, Amin MR, Kanyo R, Allison WT, Ali DW. CB 1 and CB 2 receptors play differential roles in early zebrafish locomotor development. ACTA ACUST UNITED AC 2019; 222:jeb.206680. [PMID: 31253713 DOI: 10.1242/jeb.206680] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/19/2019] [Indexed: 12/22/2022]
Abstract
Endocannabinoids (eCBs) mediate their effects through actions on several receptors, including the cannabinoid receptors CB1R and CB2R. The role played by eCBs in the development of locomotor systems is not fully understood. In this study, we investigated the roles of the eCB system in zebrafish development by pharmacologically inhibiting CB1R and CB2R (with AM251 and AM630, respectively) in either the first or second day of development. We examined the morphology of motor neurons and we determined neuromuscular outputs by quantifying the amount of swimming in 5 days post-fertilization larvae. Blocking CB2R during the first day of development resulted in gross morphological deficits and reductions in heart rate that were greater than those following treatment with the CB1R blocker AM251. Blocking CB1Rs from 0 to 24 h post-fertilization resulted in an increase in the number of secondary and tertiary branches of primary motor neurons, whereas blocking CB2Rs had the opposite effect. Both treatments manifested in reduced levels of swimming. Additionally, blocking CB1Rs resulted in greater instances of non-inflated and partially inflated swim bladders compared with AM630 treatment, suggesting that at least some of the deficits in locomotion may result from an inability to adjust buoyancy. Together, these findings indicate that the eCB system is pivotal to the development of the locomotor system in zebrafish, and that perturbations of the eCB system early in life may have detrimental effects.
Collapse
Affiliation(s)
- Md Shah Sufian
- Department of Biological Sciences, CW-405 Biological Sciences Building, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9
| | - Md Ruhul Amin
- Department of Biological Sciences, CW-405 Biological Sciences Building, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9
| | - Richard Kanyo
- Department of Biological Sciences, CW-405 Biological Sciences Building, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9.,Neuroscience and Mental Health Institute, CW-405 Biological Sciences Building, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9
| | - W Ted Allison
- Department of Biological Sciences, CW-405 Biological Sciences Building, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9.,Neuroscience and Mental Health Institute, CW-405 Biological Sciences Building, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9
| | - Declan W Ali
- Department of Biological Sciences, CW-405 Biological Sciences Building, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9 .,Neuroscience and Mental Health Institute, CW-405 Biological Sciences Building, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9.,Department of Physiology, CW-405 Biological Sciences Building, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9
| |
Collapse
|
19
|
Theodore LN, Hagedorn EJ, Cortes M, Natsuhara K, Liu SY, Perlin JR, Yang S, Daily ML, Zon LI, North TE. Distinct Roles for Matrix Metalloproteinases 2 and 9 in Embryonic Hematopoietic Stem Cell Emergence, Migration, and Niche Colonization. Stem Cell Reports 2017; 8:1226-1241. [PMID: 28416284 PMCID: PMC5425629 DOI: 10.1016/j.stemcr.2017.03.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 03/14/2017] [Accepted: 03/14/2017] [Indexed: 01/06/2023] Open
Abstract
Hematopoietic stem/progenitor cells (HSPCs) are formed during ontogeny from hemogenic endothelium in the ventral wall of the dorsal aorta (VDA). Critically, the cellular mechanism(s) allowing HSPC egress and migration to secondary niches are incompletely understood. Matrix metalloproteinases (MMPs) are inflammation-responsive proteins that regulate extracellular matrix (ECM) remodeling, cellular interactions, and signaling. Here, inhibition of vascular-associated Mmp2 function caused accumulation of fibronectin-rich ECM, retention of runx1/cmyb+ HSPCs in the VDA, and delayed caudal hematopoietic tissue (CHT) colonization; these defects were absent in fibronectin mutants, indicating that Mmp2 facilitates endothelial-to-hematopoietic transition via ECM remodeling. In contrast, Mmp9 was dispensable for HSPC budding, being instead required for proper colonization of secondary niches. Significantly, these migration defects were mimicked by overexpression and blocked by knockdown of C-X-C motif chemokine-12 (cxcl12), suggesting that Mmp9 controls CHT homeostasis through chemokine regulation. Our findings indicate Mmp2 and Mmp9 play distinct but complementary roles in developmental HSPC production and migration.
Collapse
Affiliation(s)
- Lindsay N Theodore
- Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Elliott J Hagedorn
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mauricio Cortes
- Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Kelsey Natsuhara
- Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Sarah Y Liu
- Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Julie R Perlin
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Song Yang
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Madeleine L Daily
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Leonard I Zon
- Stem Cell Program, Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Trista E North
- Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 3 Blackfan Circle, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
20
|
Rutin as a Mediator of Lipid Metabolism and Cellular Signaling Pathways Interactions in Fibroblasts Altered by UVA and UVB Radiation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4721352. [PMID: 28168010 PMCID: PMC5266866 DOI: 10.1155/2017/4721352] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/05/2016] [Indexed: 02/07/2023]
Abstract
Background. Rutin is a natural nutraceutical that is a promising compound for the prevention of UV-induced metabolic changes in skin cells. The aim of this study was to examine the effects of rutin on redox and endocannabinoid systems, as well as proinflammatory and proapoptotic processes, in UV-irradiated fibroblasts. Methods. Fibroblasts exposed to UVA and UVB radiation were treated with rutin. The activities and levels of oxidants/antioxidants and endocannabinoid system components, as well as lipid, DNA, and protein oxidation products, and the proinflammatory and pro/antiapoptotic proteins expression were measured. Results. Rutin reduced UV-induced proinflammatory response and ROS generation and enhanced the activity/levels of antioxidants (SOD, GSH-Px, vitamin E, GSH, and Trx). Rutin also normalized UV-induced Nrf2 expression. Its biological activity prevented changes in the levels of the lipid mediators: MDA, 4-HNE, and endocannabinoids, as well as the endocannabinoid receptors CB1/2, VR1, and GPR55 expression. Furthermore, rutin prevented the protein modifications (tyrosine derivatives formation in particular) and decreased the levels of the proapoptotic markers—caspase-3 and cytochrome c. Conclusion. Rutin prevents UV-induced inflammation and redox imbalance at protein and transcriptional level which favors lipid, protein, and DNA protection. In consequence rutin regulates endocannabinoid system and apoptotic balance.
Collapse
|
21
|
Kwan W, North TE. Netting Novel Regulators of Hematopoiesis and Hematologic Malignancies in Zebrafish. Curr Top Dev Biol 2017; 124:125-160. [DOI: 10.1016/bs.ctdb.2016.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
22
|
|
23
|
Robertson AL, Avagyan S, Gansner JM, Zon LI. Understanding the regulation of vertebrate hematopoiesis and blood disorders - big lessons from a small fish. FEBS Lett 2016; 590:4016-4033. [PMID: 27616157 DOI: 10.1002/1873-3468.12415] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/22/2016] [Accepted: 09/07/2016] [Indexed: 12/12/2022]
Abstract
Hematopoietic stem cells (HSCs) give rise to all differentiated blood cells. Understanding the mechanisms that regulate self-renewal and lineage specification of HSCs is key for developing treatments for many human diseases. Zebrafish have emerged as an excellent model for studying vertebrate hematopoiesis. This review will highlight the unique strengths of zebrafish and important findings that have emerged from studies of blood development and disorders using this system. We discuss recent advances in our understanding of hematopoiesis, including the origin of HSCs, molecular control of their development, and key signaling pathways involved in their regulation. We highlight significant findings from zebrafish models of blood disorders and discuss their application for investigating stem cell dysfunction in disease and for the development of new therapeutics.
Collapse
Affiliation(s)
- Anne L Robertson
- Division of Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, MA, USA
| | - Serine Avagyan
- Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, MA, USA
| | - John M Gansner
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Leonard I Zon
- Howard Hughes Medical Institute, Harvard Stem Cell Institute, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Luis TC, Tremblay CS, Manz MG, North TE, King KY, Challen GA. Inflammatory signals in HSPC development and homeostasis: Too much of a good thing? Exp Hematol 2016; 44:908-12. [PMID: 27423816 DOI: 10.1016/j.exphem.2016.06.254] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 06/22/2016] [Accepted: 06/24/2016] [Indexed: 02/08/2023]
Abstract
Hematopoietic stem cells (HSCs) reside in the bone marrow and are responsible for the lifetime maintenance of the blood and bone marrow, achieved through their differentiation into the myriad cellular components and their ability to generate additional stem cells via self-renewal. Identification of intrinsic and extrinsic factors that regulate how the HSC population is maintained over the lifespan of an organism, or those that trigger differentiation into mature hematopoietic cell types, are important goals for regenerative medicine. Recent studies have found that inflammatory signals play a role in the regulation of adult HSC homeostasis and tonic innate immune signals influence HSC development during embryogenesis. Additionally, dysregulation of inflammatory cytokines, and the consequent impact of this on hematopoietic progenitors, may be a contributing factor to the hematopoietic defects that occur during aging and in patients with bone marrow failure syndromes or blood cancers. To update recent findings on this topic, the International Society for Experimental Hematology (ISEH) organized a webinar entitled "The Role of Inflammatory Signals in Embryonic HSC Development and Adult HSC Function," which we summarize here.
Collapse
Affiliation(s)
- Tiago C Luis
- Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Headington, Oxford, United Kingdom
| | - Cedric S Tremblay
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | - Markus G Manz
- Division of Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Trista E North
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Katherine Y King
- Section of Infectious Diseases, Center for Cell and Gene Therapy, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Grant A Challen
- Section of Stem Cell Biology, Division of Oncology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO.
| |
Collapse
|
25
|
Kanz D, Konantz M, Alghisi E, North TE, Lengerke C. Endothelial-to-hematopoietic transition: Notch-ing vessels into blood. Ann N Y Acad Sci 2016; 1370:97-108. [DOI: 10.1111/nyas.13030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/21/2016] [Accepted: 01/26/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Dirk Kanz
- Department of Stem Cell and Regenerative Biology; Harvard University; Boston Massachusetts
| | - Martina Konantz
- Department of Biomedicine; University Hospital Basel; Basel Switzerland
| | - Elisa Alghisi
- Department of Biomedicine; University Hospital Basel; Basel Switzerland
| | - Trista E. North
- Beth Israel Deaconess Medical Center; Harvard Medical School; Boston Massachusetts
- Harvard Stem Cell Institute; Cambridge Massachusetts
| | - Claudia Lengerke
- Department of Biomedicine; University Hospital Basel; Basel Switzerland
- Division of Hematology; University Hospital Basel; Basel Switzerland
| |
Collapse
|
26
|
Liu LY, Alexa K, Cortes M, Schatzman-Bone S, Kim AJ, Mukhopadhyay B, Cinar R, Kunos G, North TE, Goessling W. Cannabinoid receptor signaling regulates liver development and metabolism. Development 2016; 143:609-22. [PMID: 26884397 PMCID: PMC4760316 DOI: 10.1242/dev.121731] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/05/2016] [Indexed: 12/21/2022]
Abstract
Endocannabinoid (EC) signaling mediates psychotropic effects and regulates appetite. By contrast, potential roles in organ development and embryonic energy consumption remain unknown. Here, we demonstrate that genetic or chemical inhibition of cannabinoid receptor (Cnr) activity disrupts liver development and metabolic function in zebrafish (Danio rerio), impacting hepatic differentiation, but not endodermal specification: loss of cannabinoid receptor 1 (cnr1) and cnr2 activity leads to smaller livers with fewer hepatocytes, reduced liver-specific gene expression and proliferation. Functional assays reveal abnormal biliary anatomy and lipid handling. Adult cnr2 mutants are susceptible to hepatic steatosis. Metabolomic analysis reveals reduced methionine content in Cnr mutants. Methionine supplementation rescues developmental and metabolic defects in Cnr mutant livers, suggesting a causal relationship between EC signaling, methionine deficiency and impaired liver development. The effect of Cnr on methionine metabolism is regulated by sterol regulatory element-binding transcription factors (Srebfs), as their overexpression rescues Cnr mutant liver phenotypes in a methionine-dependent manner. Our work describes a novel developmental role for EC signaling, whereby Cnr-mediated regulation of Srebfs and methionine metabolism impacts liver development and function.
Collapse
Affiliation(s)
- Leah Y Liu
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kristen Alexa
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mauricio Cortes
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | | | - Andrew J Kim
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bani Mukhopadhyay
- Laboratory of Physiological Studies, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20982, USA
| | - Resat Cinar
- Laboratory of Physiological Studies, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20982, USA
| | - George Kunos
- Laboratory of Physiological Studies, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20982, USA
| | - Trista E North
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Wolfram Goessling
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA Harvard Stem Cell Institute, Cambridge, MA 02138, USA Gastroenterology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA Dana-Farber Cancer Institute, Boston, MA 02215, USA Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|