151
|
Bonney EA, Johnson MR. The role of maternal T cell and macrophage activation in preterm birth: Cause or consequence? Placenta 2019; 79:53-61. [PMID: 30929747 DOI: 10.1016/j.placenta.2019.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/06/2019] [Indexed: 12/18/2022]
Abstract
The role of the immune system in term (TL) and preterm labor (PTL) is unknown. Despite the fact that globally, PTL remains the most important cause of childhood mortality. Infection, typically of the fetal membranes, termed chorioamnionitis, is the best-understood driver of PTL, but the mechanisms underpinning other causes, including idiopathic and stretch-induced PTL, are unclear, but may well involve activation of the maternal immune system. The final common pathway of placental dysfunction, fetal membrane rupture, cervical dilation and uterine contractions are highly complex processes. At term, choriodecidual rather than myometrial inflammation is thought to drive the onset of labor and similar findings are present in different types of PTL including idiopathic PTL. Although accumulated data has confirmed an association between the immune response and preterm birth, there is yet a need to understand if this response is an initiator or a consequence of tissue-level dysregulation. This review focuses on the potential role of macrophages and T cells in innate and adaptive immunity relevant to preterm birth in humans and animal models.
Collapse
Affiliation(s)
- Elizabeth A Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences University of Vermont, Larner College of Medicine, Burlington, VT, USA.
| | - Mark R Johnson
- Faculty of Medicine, Department of Surgery & Cancer, Imperial College, London, United Kingdom
| |
Collapse
|
152
|
Affiliation(s)
- Ai Ing Lim
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Oliver J Harrison
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA.
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA.
- Center for Human Immunology, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
153
|
Schreurs RRCE, Baumdick ME, Sagebiel AF, Kaufmann M, Mokry M, Klarenbeek PL, Schaltenberg N, Steinert FL, van Rijn JM, Drewniak A, The SMML, Bakx R, Derikx JPM, de Vries N, Corpeleijn WE, Pals ST, Gagliani N, Friese MA, Middendorp S, Nieuwenhuis EES, Reinshagen K, Geijtenbeek TBH, van Goudoever JB, Bunders MJ. Human Fetal TNF-α-Cytokine-Producing CD4 + Effector Memory T Cells Promote Intestinal Development and Mediate Inflammation Early in Life. Immunity 2019; 50:462-476.e8. [PMID: 30770246 DOI: 10.1016/j.immuni.2018.12.010] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 10/01/2018] [Accepted: 12/05/2018] [Indexed: 11/29/2022]
Abstract
Although the fetal immune system is considered tolerogenic, preterm infants can suffer from severe intestinal inflammation, including necrotizing enterocolitis (NEC). Here, we demonstrate that human fetal intestines predominantly contain tumor necrosis factor-α (TNF-α)+CD4+CD69+ T effector memory (Tem) cells. Single-cell RNA sequencing of fetal intestinal CD4+ T cells showed a T helper 1 phenotype and expression of genes mediating epithelial growth and cell cycling. Organoid co-cultures revealed a dose-dependent, TNF-α-mediated effect of fetal intestinal CD4+ T cells on intestinal stem cell (ISC) development, in which low T cell numbers supported epithelial development, whereas high numbers abrogated ISC proliferation. CD4+ Tem cell frequencies were higher in inflamed intestines from preterm infants with NEC than in healthy infant intestines and showed enhanced TNF signaling. These findings reveal a distinct population of TNF-α-producing CD4+ T cells that promote mucosal development in fetal intestines but can also mediate inflammation upon preterm birth.
Collapse
Affiliation(s)
- Renée R C E Schreurs
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Martin E Baumdick
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Adrian F Sagebiel
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Max Kaufmann
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Michal Mokry
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Paul L Klarenbeek
- Department of Clinical Immunology and Rheumatology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Nicola Schaltenberg
- Department of General, Visceral, and Thoracic Surgery and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Fenja L Steinert
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Jorik M van Rijn
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Agata Drewniak
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Kiadis Pharma B.V., Amsterdam 1105 BV, the Netherlands
| | - Sarah-May M L The
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, Amsterdam University Medical Center, Amsterdam 1105 AZ, the Netherlands
| | - Roel Bakx
- Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, Amsterdam University Medical Center, Amsterdam 1105 AZ, the Netherlands
| | - Joep P M Derikx
- Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, Amsterdam University Medical Center, Amsterdam 1105 AZ, the Netherlands
| | - Niek de Vries
- Department of Clinical Immunology and Rheumatology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Willemijn E Corpeleijn
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Steven T Pals
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Nicola Gagliani
- Department of General, Visceral, and Thoracic Surgery and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm 17176, Sweden
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Sabine Middendorp
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Edward E S Nieuwenhuis
- Division of Pediatrics, Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, Utrecht University Medical Center, Utrecht University, Utrecht 3584 EA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht University Medical Center, University of Utrecht, Utrecht 3584 CT, the Netherlands
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Johannes B van Goudoever
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam 1081 HV, the Netherlands
| | - Madeleine J Bunders
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany.
| |
Collapse
|
154
|
Fultang L, Gamble LD, Gneo L, Berry AM, Egan SA, De Bie F, Yogev O, Eden GL, Booth S, Brownhill S, Vardon A, McConville CM, Cheng PN, Norris MD, Etchevers HC, Murray J, Ziegler DS, Chesler L, Schmidt R, Burchill SA, Haber M, De Santo C, Mussai F. Macrophage-Derived IL1β and TNFα Regulate Arginine Metabolism in Neuroblastoma. Cancer Res 2019; 79:611-624. [PMID: 30545920 PMCID: PMC6420118 DOI: 10.1158/0008-5472.can-18-2139] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/19/2018] [Accepted: 12/05/2018] [Indexed: 12/18/2022]
Abstract
Neuroblastoma is the most common childhood solid tumor, yet the prognosis for high-risk disease remains poor. We demonstrate here that arginase 2 (ARG2) drives neuroblastoma cell proliferation via regulation of arginine metabolism. Targeting arginine metabolism, either by blocking cationic amino acid transporter 1 (CAT-1)-dependent arginine uptake in vitro or therapeutic depletion of arginine by pegylated recombinant arginase BCT-100, significantly delayed tumor development and prolonged murine survival. Tumor cells polarized infiltrating monocytes to an M1-macrophage phenotype, which released IL1β and TNFα in a RAC-alpha serine/threonine-protein kinase (AKT)-dependent manner. IL1β and TNFα established a feedback loop to upregulate ARG2 expression via p38 and extracellular regulated kinases 1/2 (ERK1/2) signaling in neuroblastoma and neural crest-derived cells. Proteomic analysis revealed that enrichment of IL1β and TNFα in stage IV human tumor microenvironments was associated with a worse prognosis. These data thus describe an immune-metabolic regulatory loop between tumor cells and infiltrating myeloid cells regulating ARG2, which can be clinically exploited. SIGNIFICANCE: These findings illustrate that cross-talk between myeloid cells and tumor cells creates a metabolic regulatory loop that promotes neuroblastoma progression.
Collapse
Affiliation(s)
- Livingstone Fultang
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Laura D Gamble
- Children's Cancer Institute, University of New South Wales, Sydney, Australia
| | - Luciana Gneo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Andrea M Berry
- Children's Cancer Research Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Sharon A Egan
- School of Veterinary Medicine and Science, Sutton Bonington Campus, University of Nottingham, Nottingham, UK
| | - Fenna De Bie
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Orli Yogev
- The Institute of Cancer Research, London, UK
| | - Georgina L Eden
- Children's Cancer Institute, University of New South Wales, Sydney, Australia
| | - Sarah Booth
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Samantha Brownhill
- Children's Cancer Research Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Ashley Vardon
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Carmel M McConville
- Institute of Cancer Genomic Sciences, University of Birmingham, Birmingham, Birmingham, UK
| | | | - Murray D Norris
- Children's Cancer Institute, University of New South Wales, Sydney, Australia
| | | | - Jayne Murray
- Children's Cancer Institute, University of New South Wales, Sydney, Australia
| | - David S Ziegler
- Children's Cancer Institute, University of New South Wales, Sydney, Australia
| | | | | | - Susan A Burchill
- Children's Cancer Research Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Michelle Haber
- Children's Cancer Institute, University of New South Wales, Sydney, Australia
| | - Carmela De Santo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Francis Mussai
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| |
Collapse
|
155
|
Mattar CNZ, Tan YW, Johana N, Biswas A, Tan LG, Choolani M, Bakkour S, Johnson M, Chan JKY, Flake AW. Fetoscopic versus Ultrasound-Guided Intravascular Delivery of Maternal Bone Marrow Cells in Fetal Macaques: A Technical Model for Intrauterine Haemopoietic Cell Transplantation. Fetal Diagn Ther 2019; 46:175-186. [PMID: 30661073 DOI: 10.1159/000493791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/14/2018] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Significant limitations with existing treatments for major haemoglobinopathies motivate the development of effective intrauterine therapy. We assessed the feasibility of fetoscopic and ultrasound-guided intrauterine haemopoietic cell transplantation (IUHCT) in macaque fetuses in early gestation when haemopoietic and immunological ontogeny is anticipated to enable long-term donor cell engraftment. MATERIAL AND METHODS Fluorescent-labelled bone marrow-derived mononuclear cells from 10 pregnant Macaca fascicularis were injected into their fetuses at E71-114 (18.9-170.0E+6 cells/fetus) by fetoscopic intravenous (n = 7) or ultrasound (US)-guided intracardiac injections, with sacrifice at 24 h to examine donor-cell distribution. RESULTS Operating times ranged from 35 to 118 min. Chorionic membrane tenting and intrachorionic haemorrhage were observed only with fetoscopy (n = 2). Labelled cells were stereoscopically visualised in lung, spleen, liver, and placenta. Donor-cell chimerism was highest in liver, spleen, and heart by flow cytometry, placenta by unique polymorphism qPCR, and was undetected in blood. Chimerism was 2-3 log-fold lower in individual organs by qPCR than by flow cytometry. DISCUSSION Both fetoscopic and US-guided IUHCT were technically feasible, but fetoscopy caused more intraoperative complications in our pilot series. The discrepancy in chimerism detection predicts the challenges in long-term surveillance of donor-cell chimerism. Further studies of long-term outcomes in the non-human primate are valuable for the development of clinical protocols for IUHCT.
Collapse
Affiliation(s)
- Citra N Z Mattar
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yi-Wan Tan
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Nuryanti Johana
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Arijit Biswas
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lay-Geok Tan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mahesh Choolani
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sonia Bakkour
- Blood Systems Research Institute, San Francisco, California, USA
| | - Mark Johnson
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jerry K Y Chan
- Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore, .,Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, Singapore,
| | - Alan W Flake
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
156
|
Papaioannou NE, Pasztoi M, Schraml BU. Understanding the Functional Properties of Neonatal Dendritic Cells: A Doorway to Enhance Vaccine Effectiveness? Front Immunol 2019; 9:3123. [PMID: 30687326 PMCID: PMC6335269 DOI: 10.3389/fimmu.2018.03123] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/18/2018] [Indexed: 12/31/2022] Open
Abstract
Increased susceptibility to infectious diseases is a hallmark of the neonatal period of life that is generally attributed to a relative immaturity of the immune system. Dendritic cells (DCs) are innate immune sentinels with vital roles in the initiation and orchestration of immune responses, thus, constituting a promising target for promoting neonatal immunity. However, as is the case for other immune cells, neonatal DCs have been suggested to be functionally immature compared to their adult counterparts. Here we review some of the unique aspects of neonatal DCs that shape immune responses in early life and speculate whether the functional properties of neonatal DCs could be exploited or manipulated to promote more effective vaccination in early life.
Collapse
Affiliation(s)
- Nikos E Papaioannou
- Biomedical Center, Institute for Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Maria Pasztoi
- Biomedical Center, Institute for Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Barbara U Schraml
- Biomedical Center, Institute for Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Munich, Germany.,Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
157
|
Paller AS, Kong HH, Seed P, Naik S, Scharschmidt TC, Gallo RL, Luger T, Irvine AD. The microbiome in patients with atopic dermatitis. J Allergy Clin Immunol 2018; 143:26-35. [PMID: 30476499 PMCID: PMC7163929 DOI: 10.1016/j.jaci.2018.11.015] [Citation(s) in RCA: 279] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022]
Abstract
As an interface with the environment, the skin is a complex ecosystem colonized by many microorganisms that coexist in an established balance. The cutaneous microbiome inhibits colonization with pathogens, such as Staphylococcus aureus, and is a crucial component for function of the epidermal barrier. Moreover, crosstalk between commensals and the immune system is now recognized because microorganisms can modulate both innate and adaptive immune responses. Host-commensal interactions also have an effect on the developing immune system in infants and, subsequently, the occurrence of diseases, such as asthma and atopic dermatitis (AD). Later in life, the cutaneous microbiome contributes to the development and course of skin disease. Accordingly, in patients with AD, a decrease in microbiome diversity correlates with disease severity and increased colonization with pathogenic bacteria, such as S aureus. Early clinical studies suggest that topical application of commensal organisms (eg, Staphylococcus hominis or Roseomonas mucosa) reduces AD severity, which supports an important role for commensals in decreasing S aureus colonization in patients with AD. Advancing knowledge of the cutaneous microbiome and its function in modulating the course of skin disorders, such as AD, might result in novel therapeutic strategies.
Collapse
Affiliation(s)
- Amy S Paller
- Dermatology and Infectious Disease, Northwestern University Feinberg School of Medicine and the Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, Ill.
| | - Heidi H Kong
- Dermatology Branch, National Institute of Arthritis and Musculoskletal and Skin Diseases, Bethesda, Md
| | - Patrick Seed
- Dermatology and Infectious Disease, Northwestern University Feinberg School of Medicine and the Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, Ill
| | - Shruti Naik
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY
| | | | | | - Thomas Luger
- Dermatology, University of Muenster, Munster, Germany
| | - Alan D Irvine
- Paediatric Dermatology, Our Lady's Children's Hospital Crumlin, National Children's Research Centre and Trinity College, Dublin, Ireland.
| |
Collapse
|
158
|
Oppong-Nonterah GO, Lakhdari O, Yamamura A, Hoffman HM, Prince LS. TLR Activation Alters Bone Marrow-Derived Macrophage Differentiation. J Innate Immun 2018; 11:99-108. [PMID: 30408777 DOI: 10.1159/000494070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/11/2018] [Indexed: 12/24/2022] Open
Abstract
Early exposure to inflammatory signals may have a lasting impact on immune function. Present throughout embryogenesis, macrophages are key cells providing innate immune protection to the developing fetus and newborn. Here, we have used an established model of macrophage development to test how early inflammatory signals can impact cellular differentiation and function. Bone marrow-derived macrophages were treated with Escherichia coli lipopolysaccharide (LPS) 2 days after initial isolation and culture. LPS treatment during this early stage of differentiation decreased the expression of CSF1R and increased that of the mature macrophage marker F4/80. These early changes in macrophage differentiation were also measured in cells from mice lacking IKKβ, but the change in CSF1R expression after LPS treatment was blocked with MAPK inhibition. LPS-induced changes in macrophage marker expression persisted following LPS removal, suggesting that early inflammatory activation could induce a lasting developmental impact. Early LPS exposure inhibited macrophage phagocytosis of labeled E. coli while LPS had no effect on fully differentiated macrophages. Our data demonstrate that early inflammatory exposure to a microbial stimulus induce lasting phenotypic changes in macrophages.
Collapse
Affiliation(s)
- Gertrude O Oppong-Nonterah
- Department of Pediatrics, University of California San Diego, Rady Children's Hospital, San Diego, California, USA
| | - Omar Lakhdari
- Department of Pediatrics, University of California San Diego, Rady Children's Hospital, San Diego, California, USA
| | - Asami Yamamura
- Department of Pediatrics, University of California San Diego, Rady Children's Hospital, San Diego, California, USA
| | - Hal M Hoffman
- Department of Pediatrics, University of California San Diego, Rady Children's Hospital, San Diego, California, USA
| | - Lawrence S Prince
- Department of Pediatrics, University of California San Diego, Rady Children's Hospital, San Diego, California, USA,
| |
Collapse
|
159
|
Cui L, Lu H, Lee YH. Challenges and emergent solutions for LC-MS/MS based untargeted metabolomics in diseases. MASS SPECTROMETRY REVIEWS 2018; 37:772-792. [PMID: 29486047 DOI: 10.1002/mas.21562] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 02/02/2018] [Indexed: 05/03/2023]
Abstract
In the past decade, advances in liquid chromatography-mass spectrometry (LC-MS) have revolutionized untargeted metabolomics analyses. By mining metabolomes more deeply, researchers are now primed to uncover key metabolites and their associations with diseases. The employment of untargeted metabolomics has led to new biomarker discoveries and a better mechanistic understanding of diseases with applications in precision medicine. However, many major pertinent challenges remain. First, compound identification has been poor, and left an overwhelming number of unidentified peaks. Second, partial, incomplete metabolomes persist due to factors such as limitations in mass spectrometry data acquisition speeds, wide-range of metabolites concentrations, and cellular/tissue/temporal-specific expression changes that confound our understanding of metabolite perturbations. Third, to contextualize metabolites in pathways and biology is difficult because many metabolites partake in multiple pathways, have yet to be described species specificity, or possess unannotated or more-complex functions that are not easily characterized through metabolomics analyses. From a translational perspective, information related to novel metabolite biomarkers, metabolic pathways, and drug targets might be sparser than they should be. Thankfully, significant progress has been made and novel solutions are emerging, achieved through sustained academic and industrial community efforts in terms of hardware, computational, and experimental approaches. Given the rapidly growing utility of metabolomics, this review will offer new perspectives, increase awareness of the major challenges in LC-MS metabolomics that will significantly benefit the metabolomics community and also the broader the biomedical community metabolomics aspire to serve.
Collapse
Affiliation(s)
- Liang Cui
- Translational 'Omics and Biomarkers Group, KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore
- Infectious Diseases-Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Haitao Lu
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yie Hou Lee
- Translational 'Omics and Biomarkers Group, KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore
- OBGYN-Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
160
|
Ng KP, Manjeri A, Lee LM, Chan ZE, Tan CY, Tan QD, Majeed A, Lee KL, Chuah C, Suda T, Ong ST. The arginase inhibitor Nω-hydroxy-nor-arginine (nor-NOHA) induces apoptosis in leukemic cells specifically under hypoxic conditions but CRISPR/Cas9 excludes arginase 2 (ARG2) as the functional target. PLoS One 2018; 13:e0205254. [PMID: 30307989 PMCID: PMC6181325 DOI: 10.1371/journal.pone.0205254] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/23/2018] [Indexed: 01/10/2023] Open
Abstract
Cancer cells, including in chronic myeloid leukemia (CML), depend on the hypoxic response to persist in hosts and evade therapy. Accordingly, there is significant interest in drugging cancer-specific hypoxic responses. However, a major challenge in leukemia is identifying differential and druggable hypoxic responses between leukemic and normal cells. Previously, we found that arginase 2 (ARG2), an enzyme of the urea cycle, is overexpressed in CML but not normal progenitors. ARG2 is a target of the hypoxia inducible factors (HIF1−α and HIF2−α), and is required for the generation of polyamines which are required for cell growth. We therefore explored if the clinically-tested arginase inhibitor Nω−hydroxy−nor−arginine (nor−NOHA) would be effective against leukemic cells under hypoxic conditions. Remarkably, nor−NOHA effectively induced apoptosis in ARG2-expressing cells under hypoxia but not normoxia. Co-treatment with nor−NOHA overcame hypoxia-mediated resistance towards BCR−ABL1 kinase inhibitors. While nor−NOHA itself is promising in targeting the leukemia hypoxic response, we unexpectedly found that its anti-leukemic activity was independent of ARG2 inhibition. Genetic ablation of ARG2 using CRISPR/Cas9 had no effect on the viability of leukemic cells and their sensitivity towards nor−NOHA. This discrepancy was further evidenced by the distinct effects of ARG2 knockouts and nor−NOHA on cellular respiration. In conclusion, we show that nor−NOHA has significant but off-target anti-leukemic activity among ARG2-expressing hypoxic cells. Since nor−NOHA has been employed in clinical trials, and is widely used in studies on endothelial dysfunction, immunosuppression and metabolism, the diverse biological effects of nor−NOHA must be cautiously evaluated before attributing its activity to ARG inhibition.
Collapse
Affiliation(s)
- King Pan Ng
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Aditi Manjeri
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Lin Ming Lee
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Zhu En Chan
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Chin Yee Tan
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Qiancheng Darren Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - A'Qilah Majeed
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Kian Leong Lee
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Charles Chuah
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore.,Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Toshio Suda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,International Research Center for Medical Sciences, Kumamoto University, Japan
| | - S Tiong Ong
- Cancer and Stem Cell Biology Signature Research Program, Duke-NUS Medical School, Singapore, Singapore.,Department of Haematology, Singapore General Hospital, Singapore, Singapore.,Department of Medical Oncology, National Cancer Centre, Singapore, Singapore.,Department of Medicine, Duke University Medical Center, Durham, NC, United States of America
| |
Collapse
|
161
|
Human T Cell Development, Localization, and Function throughout Life. Immunity 2018; 48:202-213. [PMID: 29466753 DOI: 10.1016/j.immuni.2018.01.007] [Citation(s) in RCA: 701] [Impact Index Per Article: 116.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/07/2017] [Accepted: 01/08/2018] [Indexed: 01/03/2023]
Abstract
Throughout life, T cells coordinate multiple aspects of adaptive immunity, including responses to pathogens, allergens, and tumors. In mouse models, the role of T cells is studied in the context of a specific type of pathogen, antigen, or disease condition over a limited time frame, whereas in humans, T cells control multiple insults simultaneously throughout the body and maintain immune homeostasis over decades. In this review, we discuss how human T cells develop and provide essential immune protection at different life stages and highlight tissue localization and subset delineation as key determinants of the T cell functional role in immune responses. We also discuss how anatomic compartments undergo distinct age-associated changes in T cell subset composition and function over a lifetime. It is important to consider age and tissue influences on human T cells when developing targeted strategies to modulate T cell-mediated immunity in vaccines and immunotherapies.
Collapse
|
162
|
Cha L, Jones AP, Trend S, Byrne SN, Fabis-Pedrini MJ, Carroll WM, Lucas RM, Cole JM, Booth DR, Kermode AG, Hart PH. Tryptophan and arginine catabolic enzymes and regulatory cytokines in clinically isolated syndrome and multiple sclerosis. Clin Transl Immunology 2018; 7:e1037. [PMID: 30128151 PMCID: PMC6095938 DOI: 10.1002/cti2.1037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/12/2018] [Accepted: 07/25/2018] [Indexed: 12/27/2022] Open
Abstract
Objectives Clinically isolated syndrome (CIS) is the earliest clinical episode in multiple sclerosis (MS). A study of circulating cells from patients with CIS may help us understand the transition to, and processes associated with, the development of MS. Methods As immune cell activity can be determined by flux through metabolic pathways, the mRNA expression of l‐tryptophan‐ and l‐arginine‐catabolising enzymes, indoleamine 2,3‐dioxygenase (IDO) 1 and IDO2 and arginase (ARG) 1 and ARG2, respectively, was compared between peripheral blood mononuclear cells (PBMCs) from healthy controls, and patients with CIS and definite MS. As one measure of cell function, cytokine mRNA levels were analysed directly ex vivo and in cells after culture for 4 h in the absence of regulatory factors in autologous serum. Results When measured directly ex vivo, the expression of IDO and ARG was greater in cells from patients with CIS and MS than cells from healthy controls. Although not linked to IDO and ARG expression, PBMCs from the CIS patients were characterised by low IL‐10 and TGFB mRNA levels and not by greater expression of proinflammatory cytokines. When the cells were cultured for 4 h without autologous serum, pro‐ and anti‐inflammatory cytokine mRNA levels positively correlated with IDO1 expression, and TGFB mRNA levels correlated with ARG1 expression. Conclusion Higher IDO and ARG expression in CIS and MS provides one sustained homeostatic mechanism to control MS‐associated inflammation. However, potent extrinsic mediators in serum may regulate immune cell function in CIS and associations between IDO, ARG and cytokine expression.
Collapse
Affiliation(s)
- Lilian Cha
- Telethon Kids Institute University of Western Australia Perth WA Australia
| | - Anderson P Jones
- Telethon Kids Institute University of Western Australia Perth WA Australia
| | - Stephanie Trend
- Telethon Kids Institute University of Western Australia Perth WA Australia
| | - Scott N Byrne
- Faculty of Medicine and Health Westmead Institute for Medical Research University of Sydney Westmead NSW Australia
| | - Marzena J Fabis-Pedrini
- Centre for Neuromuscular and Neurological Disorders Perron Institute for Neurological and Translational Science Sir Charles Gairdner Hospital University of Western Australia Perth WA Australia
| | - William M Carroll
- Centre for Neuromuscular and Neurological Disorders Perron Institute for Neurological and Translational Science Sir Charles Gairdner Hospital University of Western Australia Perth WA Australia
| | - Robyn M Lucas
- National Centre for Epidemiology and Population Health Research School of Population Health Australian National University Canberra ACT Australia
| | | | - David R Booth
- Faculty of Medicine and Health Westmead Institute for Medical Research University of Sydney Westmead NSW Australia
| | - Allan G Kermode
- Centre for Neuromuscular and Neurological Disorders Perron Institute for Neurological and Translational Science Sir Charles Gairdner Hospital University of Western Australia Perth WA Australia.,Institute for Immunology and Infectious Disease Murdoch University Perth WA Australia
| | - Prue H Hart
- Telethon Kids Institute University of Western Australia Perth WA Australia
| |
Collapse
|
163
|
Jassinskaja M, Johansson E, Kristiansen TA, Åkerstrand H, Sjöholm K, Hauri S, Malmström J, Yuan J, Hansson J. Comprehensive Proteomic Characterization of Ontogenic Changes in Hematopoietic Stem and Progenitor Cells. Cell Rep 2018; 21:3285-3297. [PMID: 29241553 DOI: 10.1016/j.celrep.2017.11.070] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/27/2017] [Accepted: 11/19/2017] [Indexed: 12/22/2022] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) in the fetus and adult possess distinct molecular landscapes that regulate cell fate and change their susceptibility to initiation and progression of hematopoietic malignancies. Here, we applied in-depth quantitative proteomics to comprehensively describe and compare the proteome of fetal and adult HSPCs. Our data uncover a striking difference in complexity of the cellular proteomes, with more diverse adult-specific HSPC proteomic signatures. The differential protein content in fetal and adult HSPCs indicate distinct metabolic profiles and protein complex stoichiometries. Additionally, adult characteristics include an arsenal of proteins linked to viral and bacterial defense, as well as protection against ROS-induced protein oxidation. Further analyses show that interferon α, as well as Neutrophil elastase, has distinct functional effects in fetal and adult HSPCs. This study provides a rich resource aimed toward an enhanced mechanistic understanding of normal and malignant hematopoiesis during fetal and adult life.
Collapse
Affiliation(s)
- Maria Jassinskaja
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden
| | - Emil Johansson
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden
| | - Trine Ahn Kristiansen
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden
| | - Hugo Åkerstrand
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden
| | - Kristoffer Sjöholm
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, 221 84 Lund, Sweden
| | - Simon Hauri
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, 221 84 Lund, Sweden
| | - Johan Malmström
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, 221 84 Lund, Sweden
| | - Joan Yuan
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden
| | - Jenny Hansson
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden.
| |
Collapse
|
164
|
Delyea C, Bozorgmehr N, Koleva P, Dunsmore G, Shahbaz S, Huang V, Elahi S. CD71 + Erythroid Suppressor Cells Promote Fetomaternal Tolerance through Arginase-2 and PDL-1. THE JOURNAL OF IMMUNOLOGY 2018; 200:4044-4058. [PMID: 29735482 DOI: 10.4049/jimmunol.1800113] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/16/2018] [Indexed: 12/31/2022]
Abstract
Survival of the allogeneic pregnancy depends on the maintenance of immune tolerance to paternal alloantigens at the fetomaternal interface. Multiple localized mechanisms contribute to the fetal evasion from the mother's immune rejection as the fetus is exposed to a wide range of stimulatory substances such as maternal alloantigens, microbes and amniotic fluids. In this article, we demonstrate that CD71+ erythroid cells are expanded at the fetomaternal interface and in the periphery during pregnancy in both humans and mice. These cells exhibit immunosuppressive properties, and their abundance is associated with a Th2 skewed immune response, as their depletion results in a proinflammatory immune response at the fetomaternal interface. In addition to their function in suppressing proinflammatory responses in vitro, maternal CD71+ erythroid cells inhibit an aggressive allogeneic response directed against the fetus such as reduction in TNF-α and IFN-γ production through arginase-2 activity and PD-1/programmed death ligand-1 (PDL-1) interactions. Their depletion leads to the failure of gestation due to the immunological rejection of the fetus. Similarly, fetal liver CD71+ erythroid cells exhibit immunosuppressive activity. Therefore, immunosuppression mediated by CD71+ erythroid cells on both sides (mother/fetus) is crucial for fetomaternal tolerance. Thus, our results reveal a previously unappreciated role for CD71+ erythroid cells in pregnancy and indicate that these cells mediate homeostatic immunosuppressive/immunoregulatory responses during pregnancy.
Collapse
Affiliation(s)
- Cole Delyea
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2E1 Alberta, Canada
| | - Najmeh Bozorgmehr
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2E1 Alberta, Canada
| | - Petya Koleva
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2E1 Alberta, Canada
| | - Garett Dunsmore
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2E1 Alberta, Canada.,Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2E1 Alberta, Canada
| | - Shima Shahbaz
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2E1 Alberta, Canada
| | - Vivian Huang
- Division of Gastroenterology, University of Alberta, Edmonton, T6G 2E1 Alberta, Canada; and.,Division of Gastroenterology, University of Toronto, Mount Sinai Hospital, Sinai Health System, Toronto, M5G 1X5 Ontario, Canada
| | - Shokrollah Elahi
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2E1 Alberta, Canada; .,Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2E1 Alberta, Canada
| |
Collapse
|
165
|
Collin M, Bigley V. Human dendritic cell subsets: an update. Immunology 2018; 154:3-20. [PMID: 29313948 PMCID: PMC5904714 DOI: 10.1111/imm.12888] [Citation(s) in RCA: 808] [Impact Index Per Article: 134.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/30/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023] Open
Abstract
Dendritic cells (DC) are a class of bone-marrow-derived cells arising from lympho-myeloid haematopoiesis that form an essential interface between the innate sensing of pathogens and the activation of adaptive immunity. This task requires a wide range of mechanisms and responses, which are divided between three major DC subsets: plasmacytoid DC (pDC), myeloid/conventional DC1 (cDC1) and myeloid/conventional DC2 (cDC2). Each DC subset develops under the control of a specific repertoire of transcription factors involving differential levels of IRF8 and IRF4 in collaboration with PU.1, ID2, E2-2, ZEB2, KLF4, IKZF1 and BATF3. DC haematopoiesis is conserved between mammalian species and is distinct from monocyte development. Although monocytes can differentiate into DC, especially during inflammation, most quiescent tissues contain significant resident populations of DC lineage cells. An extended range of surface markers facilitates the identification of specific DC subsets although it remains difficult to dissociate cDC2 from monocyte-derived DC in some settings. Recent studies based on an increasing level of resolution of phenotype and gene expression have identified pre-DC in human blood and heterogeneity among cDC2. These advances facilitate the integration of mouse and human immunology, support efforts to unravel human DC function in vivo and continue to present new translational opportunities to medicine.
Collapse
Affiliation(s)
- Matthew Collin
- Human Dendritic Cell LabInstitute of Cellular Medicine and NIHR Newcastle Biomedical Research Centre Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle UniversityNewcastle upon TyneUK
| | - Venetia Bigley
- Human Dendritic Cell LabInstitute of Cellular Medicine and NIHR Newcastle Biomedical Research Centre Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
166
|
Sharma A, Rudra D. Emerging Functions of Regulatory T Cells in Tissue Homeostasis. Front Immunol 2018; 9:883. [PMID: 29887862 PMCID: PMC5989423 DOI: 10.3389/fimmu.2018.00883] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/10/2018] [Indexed: 01/12/2023] Open
Abstract
CD4+Foxp3+ regulatory T-cells (Tregs) are a unique subset of helper T-cells, which regulate immune response and establish peripheral tolerance. Tregs not only maintain the tone and tenor of an immune response by dominant tolerance but, in recent years, have also been identified as key players in resolving tissue inflammation and as mediators of tissue healing. Apart from being diverse in their origin (thymic and peripheral) and location (lymphoid and tissue resident), Tregs are also phenotypically heterogeneous as per the orientation of ongoing immune response. In this review, we discuss the recent advances in the field of Treg biology in general, and non-lymphoid and tissue-resident Tregs in particular. We elaborate upon well-known visceral adipose tissue, colon, skin, and tumor-infiltrating Tregs and newly identified tissue Treg populations as in lungs, skeletal muscle, placenta, and other tissues. Our attempt is to differentiate Tregs based on distinctive properties of their location, origin, ligand specificity, chemotaxis, and specific suppressive mechanisms. Despite ever expanding roles in maintaining systemic homeostasis, Tregs are employed by large varieties of tumors to dampen antitumor immunity. Thus, a comprehensive understanding of Treg biology in the context of inflammation can be instrumental in effectively managing tissue transplantation, autoimmunity, and antitumor immune responses.
Collapse
Affiliation(s)
- Amit Sharma
- Academy of Immunology and Microbiology, Institute for Basic Science (IBS), Pohang, South Korea.,Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Dipayan Rudra
- Academy of Immunology and Microbiology, Institute for Basic Science (IBS), Pohang, South Korea.,Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| |
Collapse
|
167
|
Simoni Y, Chng MHY, Li S, Fehlings M, Newell EW. Mass cytometry: a powerful tool for dissecting the immune landscape. Curr Opin Immunol 2018; 51:187-196. [PMID: 29655022 DOI: 10.1016/j.coi.2018.03.023] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 03/25/2018] [Accepted: 03/26/2018] [Indexed: 10/17/2022]
Abstract
Advancement in methodologies for single cell analysis has historically been a major driver of progress in immunology. Currently, high dimensional flow cytometry, mass cytometry and various forms of single cell sequencing-based analysis methods are being widely adopted to expose the staggering heterogeneity of immune cells in many contexts. Here, we focus on mass cytometry, a form of flow cytometry that allows for simultaneous interrogation of more than 40 different marker molecules, including cytokines and transcription factors, without the need for spectral compensation. We argue that mass cytometry occupies an important niche within the landscape of single-cell analysis platforms that enables the efficient and in-depth study of diverse immune cell subsets with an ability to zoom-in on myeloid and lymphoid compartments in various tissues in health and disease. We further discuss the unique features of mass cytometry that are favorable for combining multiplex peptide-MHC multimer technology and phenotypic characterization of antigen specific T cells. By referring to recent studies revealing the complexities of tumor immune infiltrates, we highlight the particular importance of this technology for studying cancer in the context of cancer immunotherapy. Finally, we provide thoughts on current technical limitations and how we imagine these being overcome.
Collapse
Affiliation(s)
- Yannick Simoni
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | - Melissa Hui Yen Chng
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | - Shamin Li
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | | | - Evan W Newell
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore.
| |
Collapse
|
168
|
Brown ME, Zhou Y, McIntosh BE, Norman IG, Lou HE, Biermann M, Sullivan JA, Kamp TJ, Thomson JA, Anagnostopoulos PV, Burlingham WJ. A Humanized Mouse Model Generated Using Surplus Neonatal Tissue. Stem Cell Reports 2018; 10:1175-1183. [PMID: 29576539 PMCID: PMC5998340 DOI: 10.1016/j.stemcr.2018.02.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/20/2018] [Accepted: 02/23/2018] [Indexed: 12/30/2022] Open
Abstract
Here, we describe the NeoThy humanized mouse model created using non-fetal human tissue sources, cryopreserved neonatal thymus and umbilical cord blood hematopoietic stem cells (HSCs). Conventional humanized mouse models are made by engrafting human fetal thymus and HSCs into immunocompromised mice. These mice harbor functional human T cells that have matured in the presence of human self-peptides and human leukocyte antigen molecules. Neonatal thymus tissue is more abundant and developmentally mature and allows for creation of up to ∼50-fold more mice per donor compared with fetal tissue models. The NeoThy has equivalent frequencies of engrafted human immune cells compared with fetal tissue humanized mice and exhibits T cell function in assays of ex vivo cell proliferation, interferon γ secretion, and in vivo graft infiltration. The NeoThy model may provide significant advantages for induced pluripotent stem cell immunogenicity studies, while bypassing the requirement for fetal tissue. Neonatal tissue is a viable alternative to fetal for mouse humanization Over 1,000 NeoThy mice can be made from one neonatal thymus donor The NeoThy enables robust pre-clinical immunogenicity studies of iPSC therapies
Collapse
Affiliation(s)
- Matthew E Brown
- Division of Transplantation/Department of Surgery, University of Wisconsin, Madison, WI 53792, USA; Regenerative Biology, Morgridge Institute for Research, Madison, WI 53715, USA
| | - Ying Zhou
- Division of Transplantation/Department of Surgery, University of Wisconsin, Madison, WI 53792, USA
| | | | - Ian G Norman
- Division of Transplantation/Department of Surgery, University of Wisconsin, Madison, WI 53792, USA
| | - Hannah E Lou
- Division of Transplantation/Department of Surgery, University of Wisconsin, Madison, WI 53792, USA
| | - Mitch Biermann
- Department of Medicine, University of Wisconsin, Madison, WI 53792, USA
| | - Jeremy A Sullivan
- Division of Transplantation/Department of Surgery, University of Wisconsin, Madison, WI 53792, USA
| | - Timothy J Kamp
- Department of Medicine, University of Wisconsin, Madison, WI 53792, USA; Department of Cell & Regenerative Biology, University of Wisconsin, Madison, WI 53792, USA
| | - James A Thomson
- Regenerative Biology, Morgridge Institute for Research, Madison, WI 53715, USA; Department of Cell & Regenerative Biology, University of Wisconsin, Madison, WI 53792, USA; Department of Molecular, Cellular, & Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Petros V Anagnostopoulos
- Division of Cardiothoracic Surgery/Department of Surgery, University of Wisconsin, Madison, WI 53792, USA
| | - William J Burlingham
- Division of Transplantation/Department of Surgery, University of Wisconsin, Madison, WI 53792, USA.
| |
Collapse
|
169
|
Schultze JL, Aschenbrenner AC. Systems immunology allows a new view on human dendritic cells. Semin Cell Dev Biol 2018; 86:15-23. [PMID: 29448068 DOI: 10.1016/j.semcdb.2018.02.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 11/23/2017] [Accepted: 02/10/2018] [Indexed: 01/12/2023]
Abstract
As the most important antigen-presenting cells, dendritic cells connect the innate and adaptive part of our immune system and play a pivotal role in our course of action against invading pathogens as well as during successful vaccination. Immunologists have therefore studied these cells in great detail using flow cytometry-based analyses, in vitro assays and in vivo models, both in murine models and in humans. Albeit, sophisticated, classical immunological, and molecular approaches were often unable to unequivocally determine the subpopulation structure of the dendritic cell lineage and not surprisingly, conflicting results about dendritic cell subsets co-existed throughout the last decades. With the advent of systems approaches and the most recent introduction of -omics approaches on the single cell level combined with multi-colour flow cytometry or mass cytometry, we now enter an era allowing us to define cell population structures with an unprecedented precision. We will report here on the most recent studies applying these technologies to human dendritic cells. Proper delineation of and definition of molecular signatures for the different human dendritic cell subsets will greatly facilitate studying these cells in the future: understanding their function under physiological as well as pathological conditions.
Collapse
Affiliation(s)
- Joachim L Schultze
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Sigmund-Freud-Str. 27, 53175 Bonn, Germany.
| | - Anna C Aschenbrenner
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany.
| |
Collapse
|
170
|
Franco A, Kumar J, Lin G, Behnamfar N, Hsieh LE, Shimizu C, Tremoulet AH, Burns JC, Linden J. Pediatric tolerogenic DCs expressing CD4 and immunoglobulin-like transcript receptor (ILT)-4 secrete IL-10 in response to Fc and adenosine. Eur J Immunol 2018; 48:482-491. [PMID: 29244203 DOI: 10.1002/eji.201747139] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 10/05/2017] [Accepted: 12/11/2017] [Indexed: 11/08/2022]
Abstract
We characterized a novel population of tolerogenic myeloid dendritic cells (tmDCs) defined as CD11c+ CD11b+ CD14+ CD4+ and immunoglobulin-like transcript receptor (ILT)-4+ that are significantly more abundant in the circulation of infants and young children than in adults. TmDCs secrete the immunosuppressive lymphokine interleukin (IL)-10 when stimulated with the heavy constant region of immunoglobulins (Fc) and express high levels of the adenosine A2A receptor (A2A R), which, when activated by adenosine, inhibits the release of pro-inflammatory cytokines from most immune cells. Here we show that stimulation of the A2A R on tmDCs by regadenoson or N-ethylcarboxamidoadenosine (NECA) rapidly increases cyclic AMP accumulation and enhances IL-10 production under Fc stimulatory conditions. In co-culture experiments, tmDCs inhibit the differentiation of naïve T cells to a pro-inflammatory phenotype. In conclusion, although DCs are classically viewed as antigen presenting cells that activate T cells, we show an independent role of tmDCs in pediatric immune regulation that may be important for suppressing T cell responses to neoantigens in infants and young children.
Collapse
Affiliation(s)
- Alessandra Franco
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, University of California San Diego, School of Medicine and Rady Children's Hospital, La Jolla, CA, USA
| | - Jeetendra Kumar
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, University of California San Diego, School of Medicine and Rady Children's Hospital, La Jolla, CA, USA
| | - Gene Lin
- La Jolla Institute for Allergy and Immunology, Division of Developmental Immunology, Athena Circle, La Jolla, CA, USA
| | - Negar Behnamfar
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, University of California San Diego, School of Medicine and Rady Children's Hospital, La Jolla, CA, USA
| | - Li-En Hsieh
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, University of California San Diego, School of Medicine and Rady Children's Hospital, La Jolla, CA, USA
| | - Chisato Shimizu
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, University of California San Diego, School of Medicine and Rady Children's Hospital, La Jolla, CA, USA
| | - Adriana H Tremoulet
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, University of California San Diego, School of Medicine and Rady Children's Hospital, La Jolla, CA, USA
| | - Jane C Burns
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, University of California San Diego, School of Medicine and Rady Children's Hospital, La Jolla, CA, USA
| | - Joel Linden
- La Jolla Institute for Allergy and Immunology, Division of Developmental Immunology, Athena Circle, La Jolla, CA, USA.,Department of Pharmacology, University of California San Diego, School of Medicine, University of California, San Diego, USA
| |
Collapse
|
171
|
Sachamitr P, Leishman AJ, Davies TJ, Fairchild PJ. Directed Differentiation of Human Induced Pluripotent Stem Cells into Dendritic Cells Displaying Tolerogenic Properties and Resembling the CD141 + Subset. Front Immunol 2018; 8:1935. [PMID: 29358940 PMCID: PMC5766641 DOI: 10.3389/fimmu.2017.01935] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/15/2017] [Indexed: 12/24/2022] Open
Abstract
The advent of induced pluripotent stem cells (iPSCs) has begun to revolutionize cell therapy by providing a convenient source of rare cell types not normally available from patients in sufficient numbers for therapeutic purposes. In particular, the development of protocols for the differentiation of populations of leukocytes as diverse as naïve T cells, macrophages, and natural killer cells provides opportunities for their scale-up and quality control prior to administration. One population of leukocytes whose therapeutic potential has yet to be explored is the subset of conventional dendritic cells (DCs) defined by their surface expression of CD141. While these cells stimulate cytotoxic T cells in response to inflammation through the cross-presentation of viral and tumor-associated antigens in an MHC class I-restricted manner, under steady-state conditions CD141+ DCs resident in interstitial tissues are focused on the maintenance of homeostasis through the induction of tolerance to local antigens. Here, we describe protocols for the directed differentiation of human iPSCs into a mixed population of CD11c+ DCs through the spontaneous formation of embryoid bodies and exposure to a cocktail of growth factors, the scheduled withdrawal of which serves to guide the process of differentiation. Furthermore, we describe the enrichment of DCs expressing CD141 through depletion of CD1c+ cells, thereby obtaining a population of “untouched” DCs unaffected by cross-linking of surface CD141. The resulting cells display characteristic phagocytic and endocytic capacity and acquire an immunostimulatory phenotype following exposure to inflammatory cytokines and toll-like receptor agonists. Nevertheless, under steady-state conditions, these cells share some of the tolerogenic properties of tissue-resident CD141+ DCs, which may be further reinforced by exposure to a range of pharmacological agents including interleukin-10, rapamycin, dexamethasone, and 1α,25-dihydoxyvitamin D3. Our protocols therefore provide access to a novel source of DCs analogous to the CD141+ subset under steady-state conditions in vivo and may, therefore, find utility in the treatment of a range of disease states requiring the establishment of immunological tolerance.
Collapse
Affiliation(s)
- Patty Sachamitr
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Alison J Leishman
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Timothy J Davies
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Paul J Fairchild
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
172
|
Brizić I, Hiršl L, Britt WJ, Krmpotić A, Jonjić S. Immune responses to congenital cytomegalovirus infection. Microbes Infect 2017; 20:543-551. [PMID: 29287989 DOI: 10.1016/j.micinf.2017.12.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/15/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022]
Abstract
Human cytomegalovirus (HCMV) is the most common cause of viral infection acquired in utero. Even though the infection has been studied for several decades, immune determinants important for virus control and mechanisms of long-term sequelae caused by infection are still insufficiently characterized. Animal models of congenital HCMV infection provide unique opportunity to study various aspects of human disease. In this review, we summarize current knowledge on the role of immune system in congenital CMV infection, with emphasis on lessons learned from mouse model of congenital CMV infection.
Collapse
Affiliation(s)
- Ilija Brizić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia; Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Lea Hiršl
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia; Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - William J Britt
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pediatrics Infectious Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Astrid Krmpotić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia; Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
173
|
Steggerda SM, Bennett MK, Chen J, Emberley E, Huang T, Janes JR, Li W, MacKinnon AL, Makkouk A, Marguier G, Murray PJ, Neou S, Pan A, Parlati F, Rodriguez MLM, Van de Velde LA, Wang T, Works M, Zhang J, Zhang W, Gross MI. Inhibition of arginase by CB-1158 blocks myeloid cell-mediated immune suppression in the tumor microenvironment. J Immunother Cancer 2017; 5:101. [PMID: 29254508 PMCID: PMC5735564 DOI: 10.1186/s40425-017-0308-4] [Citation(s) in RCA: 294] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/30/2017] [Indexed: 02/08/2023] Open
Abstract
Background Myeloid cells are an abundant leukocyte in many types of tumors and contribute to immune evasion. Expression of the enzyme arginase 1 (Arg1) is a defining feature of immunosuppressive myeloid cells and leads to depletion of L-arginine, a nutrient required for T cell and natural killer (NK) cell proliferation. Here we use CB-1158, a potent and orally-bioavailable small-molecule inhibitor of arginase, to investigate the role of Arg1 in regulating anti-tumor immunity. Methods CB-1158 was tested for the ability to block myeloid cell-mediated inhibition of T cell proliferation in vitro, and for tumor growth inhibition in syngeneic mouse models of cancer as a single agent and in combination with other therapies. Tumors from animals treated with CB-1158 were profiled for changes in immune cell subsets, expression of immune-related genes, and cytokines. Human tumor tissue microarrays were probed for Arg1 expression by immunohistochemistry and immunofluorescence. Cancer patient plasma samples were assessed for Arg1 protein and L-arginine by ELISA and mass spectrometry, respectively. Results CB-1158 blocked myeloid cell-mediated suppression of T cell proliferation in vitro and reduced tumor growth in multiple mouse models of cancer, as a single agent and in combination with checkpoint blockade, adoptive T cell therapy, adoptive NK cell therapy, and the chemotherapy agent gemcitabine. Profiling of the tumor microenvironment revealed that CB-1158 increased tumor-infiltrating CD8+ T cells and NK cells, inflammatory cytokines, and expression of interferon-inducible genes. Patient tumor samples from multiple histologies expressed an abundance of tumor-infiltrating Arg1+ myeloid cells. Plasma samples from cancer patients exhibited elevated Arg1 and reduced L-arginine compared to healthy volunteers. Conclusions These results demonstrate that Arg1 is a key mediator of immune suppression and that inhibiting Arg1 with CB-1158 shifts the immune landscape toward a pro-inflammatory environment, blunting myeloid cell-mediated immune evasion and reducing tumor growth. Furthermore, our results suggest that arginase blockade by CB-1158 may be an effective therapy in multiple types of cancer and combining CB-1158 with standard-of-care chemotherapy or other immunotherapies may yield improved clinical responses.
Collapse
Affiliation(s)
- Susanne M Steggerda
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA.
| | - Mark K Bennett
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Jason Chen
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Ethan Emberley
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Tony Huang
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Julie R Janes
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Weiqun Li
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Andrew L MacKinnon
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Amani Makkouk
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Gisele Marguier
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Peter J Murray
- Max Planck Institute for Biochemistry, Martinsried, Germany.,Departments of Infectious Diseases and Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Silinda Neou
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Alison Pan
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Francesco Parlati
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Mirna L M Rodriguez
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Lee-Ann Van de Velde
- Departments of Infectious Diseases and Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Tracy Wang
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Melissa Works
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Jing Zhang
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Winter Zhang
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Matthew I Gross
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| |
Collapse
|
174
|
Freitas-Lopes MA, Mafra K, David BA, Carvalho-Gontijo R, Menezes GB. Differential Location and Distribution of Hepatic Immune Cells. Cells 2017; 6:cells6040048. [PMID: 29215603 PMCID: PMC5755505 DOI: 10.3390/cells6040048] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/03/2017] [Accepted: 12/04/2017] [Indexed: 12/12/2022] Open
Abstract
The liver is one of the main organs in the body, performing several metabolic and immunological functions that are indispensable to the organism. The liver is strategically positioned in the abdominal cavity between the intestine and the systemic circulation. Due to its location, the liver is continually exposed to nutritional insults, microbiota products from the intestinal tract, and to toxic substances. Hepatocytes are the major functional constituents of the hepatic lobes, and perform most of the liver’s secretory and synthesizing functions, although another important cell population sustains the vitality of the organ: the hepatic immune cells. Liver immune cells play a fundamental role in host immune responses and exquisite mechanisms are necessary to govern the density and the location of the different hepatic leukocytes. Here we discuss the location of these pivotal cells within the different liver compartments, and how their frequency and tissular location can dictate the fate of liver immune responses.
Collapse
Affiliation(s)
- Maria Alice Freitas-Lopes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Kassiana Mafra
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Bruna A David
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, University of Calgary. Calgary, AB T2N 1N4, Canada.
| | - Raquel Carvalho-Gontijo
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Gustavo B Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| |
Collapse
|
175
|
Jenmalm MC. The mother-offspring dyad: microbial transmission, immune interactions and allergy development. J Intern Med 2017; 282:484-495. [PMID: 28727206 DOI: 10.1111/joim.12652] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The increasing prevalence of allergy in affluent countries may be caused by reduced intensity and diversity of microbial stimulation, resulting in abnormal postnatal immune maturation. Most studies investigating the underlying immunomodulatory mechanisms have focused on postnatal microbial exposure, for example demonstrating that the gut microbiota differs in composition and diversity during the first months of life in children who later do or do not develop allergic disease. However, it is also becoming increasingly evident that the maternal microbial environment during pregnancy is important in childhood immune programming, and the first microbial encounters may occur already in utero. During pregnancy, there is a close immunological interaction between the mother and her offspring, which provides important opportunities for the maternal microbial environment to influence the immune development of the child. In support of this theory, combined pre- and postnatal supplementations seem to be crucial for the preventive effect of probiotics on infant eczema. Here, the influence of microbial and immune interactions within the mother-offspring dyad on childhood allergy development will be discussed. In addition, how perinatal transmission of microbes and immunomodulatory factors from mother to offspring may shape appropriate immune maturation during infancy and beyond, potentially via epigenetic mechanisms, will be examined. Deeper understanding of these interactions between the maternal and offspring microbiome and immunity is needed to identify efficacious preventive measures to combat the allergy epidemic.
Collapse
Affiliation(s)
- M C Jenmalm
- Department of Clinical and Experimental Medicine, Unit of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden.,International Inflammation (in-FLAME) Network of the World Universities Network, Sydney, NSW, Australia
| |
Collapse
|
176
|
Le TP, Thai TH. The State of Cellular Adoptive Immunotherapy for Neuroblastoma and Other Pediatric Solid Tumors. Front Immunol 2017; 8:1640. [PMID: 29225605 PMCID: PMC5705544 DOI: 10.3389/fimmu.2017.01640] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/09/2017] [Indexed: 01/21/2023] Open
Abstract
Research on adult cancer immunotherapy is proceeding at a rapid pace resulting in an impressive success rate exemplified by a few high profile cases. However, this momentum is not readily extended to pediatric immunotherapy, and it is not for lack of trying. Though reasons for the slower advance are not apparent, some issues can be raised. Pediatric cancer patients represent a distinct demographic group whose immune system is inherently different from that of mature adults. Treating pediatric patients with immunotherapy designed for adults may not yield objective clinical responses. Here, we will present an update on adoptive T-cell and natural killer-cell therapies for neuroblastoma and other childhood solid tumors. Additionally, we will delineate key differences between human fetal/neonatal and adult immune systems. We hope this will generate interests leading to the discussion of potential future directions for improving adoptive cancer immunotherapy for children.
Collapse
Affiliation(s)
- Thanh-Phuong Le
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - To-Ha Thai
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
177
|
Antunes MM, Carvalho ÉD, Menezes GB. DIY: "Do Imaging Yourself" - Conventional microscopes as powerful tools for in vivo investigation. Int J Biochem Cell Biol 2017; 94:1-5. [PMID: 29128683 DOI: 10.1016/j.biocel.2017.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 01/29/2023]
Abstract
Intravital imaging has been increasingly employed in cell biology studies and it is becoming one of the most powerful tools for in vivo investigation. Although some protocols can be extremely complex, most intravital imaging procedures can be performed using basic surgery and animal maintenance techniques. More importantly, regular confocal microscopes - the same that are used for imaging immunofluorescence slides - can also acquire high quality intravital images and movies after minor adaptations. Here we propose minimal adaptations in stock microscopes that allow major improvements in different fields of scientific investigation.
Collapse
Affiliation(s)
- Maísa Mota Antunes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Érika de Carvalho
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
178
|
Jennewein MF, Abu-Raya B, Jiang Y, Alter G, Marchant A. Transfer of maternal immunity and programming of the newborn immune system. Semin Immunopathol 2017; 39:605-613. [PMID: 28971246 DOI: 10.1007/s00281-017-0653-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/05/2017] [Indexed: 12/20/2022]
Abstract
As placental mammals, the pregnant women and the fetus have intense and prolonged interactions during gestation. There is increasing evidence that multiple molecular as well as cellular components originating in pregnant women are transferred to the fetus. The transfer of maternal antibodies has long been recognized as a central component of newborn immunity against pathogens. More recent studies indicate that inflammatory mediators, micronutrients, microbial products and maternal cells are transferred in utero and influence the fetal immune system. Together, these multiple signals are likely to form a complex network of interactions that program the neonatal immune system and tune its homeostatic regulation. Maternal disorders, in particular infectious diseases, modify these signals and may thereby alter immunity in early life. Understanding maternal programming of the newborn immune system could provide a basis for interventions promoting child health.
Collapse
Affiliation(s)
| | - Bahaa Abu-Raya
- Vaccine Evaluation Center, BC Children's Hospital, Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| | - Yiwei Jiang
- Institute for Medical Immunology, Université Libre de Bruxelles, Rue Adrienne Bolland 8, 6041 Gosselies, Charleroi, Belgium
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Rue Adrienne Bolland 8, 6041 Gosselies, Charleroi, Belgium.
| |
Collapse
|
179
|
Dantoft W, Martínez-Vicente P, Jafali J, Pérez-Martínez L, Martin K, Kotzamanis K, Craigon M, Auer M, Young NT, Walsh P, Marchant A, Angulo A, Forster T, Ghazal P. Genomic Programming of Human Neonatal Dendritic Cells in Congenital Systemic and In Vitro Cytomegalovirus Infection Reveal Plastic and Robust Immune Pathway Biology Responses. Front Immunol 2017; 8:1146. [PMID: 28993767 PMCID: PMC5622154 DOI: 10.3389/fimmu.2017.01146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/30/2017] [Indexed: 12/12/2022] Open
Abstract
Neonates and especially premature infants are highly susceptible to infection but still can have a remarkable resilience that is poorly understood. The view that neonates have an incomplete or deficient immune system is changing. Human neonatal studies are challenging, and elucidating host protective responses and underlying cognate pathway biology, in the context of viral infection in early life, remains to be fully explored. In both resource rich and poor settings, human cytomegalovirus (HCMV) is the most common cause of congenital infection. By using unbiased systems analyses of transcriptomic resources for HCMV neonatal infection, we find the systemic response of a preterm congenital HCMV infection, involves a focused IFN regulatory response associated with dendritic cells. Further analysis of transcriptional-programming of neonatal dendritic cells in response to HCMV infection in culture revealed an early dominant IFN-chemokine regulatory subnetworks, and at later times the plasticity of pathways implicated in cell-cycle control and lipid metabolism. Further, we identify previously unknown suppressed networks associated with infection, including a select group of GPCRs. Functional siRNA viral growth screen targeting 516-GPCRs and subsequent validation identified novel GPCR-dependent antiviral (ADORA1) and proviral (GPR146, RGS16, PTAFR, SCTR, GPR84, GPR85, NMUR2, FZ10, RDS, CCL17, and SORT1) roles. By contrast a gene family cluster of protocadherins is significantly differentially induced in neonatal cells, suggestive of possible immunomodulatory roles. Unexpectedly, programming responses of adult and neonatal dendritic cells, upon HCMV infection, demonstrated comparable quantitative and qualitative responses showing that functionally, neonatal dendritic cell are not overly compromised. However, a delay in responses of neonatal cells for IFN subnetworks in comparison with adult-derived cells are notable, suggestive of subtle plasticity differences. These findings support a set-point control mechanism rather than immaturity for explaining not only neonatal susceptibility but also resilience to infection. In summary, our findings show that neonatal HCMV infection leads to a highly plastic and functional robust programming of dendritic cells in vivo and in vitro. In comparison with adults, a minimal number of subtle quantitative and temporal differences may contribute to variability in host susceptibility and resilience, in a context dependent manner.
Collapse
Affiliation(s)
- Widad Dantoft
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Pablo Martínez-Vicente
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - James Jafali
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Lara Pérez-Martínez
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,Quantitative Proteomics, Institute of Molecular Biology, Mainz, Germany
| | - Kim Martin
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,Synexa Life Sciences, Cape Town, South Africa
| | - Konstantinos Kotzamanis
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Marie Craigon
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Manfred Auer
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom.,SynthSys-Centre for Synthetic and Systems Biology, School of Engineering, University of Edinburgh, Edinburgh, United Kingdom
| | - Neil T Young
- Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Paul Walsh
- NSilico Life Science and Department of Computing, Institute of Technology, Cork, Ireland
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Charleroi, Belgium
| | - Ana Angulo
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Thorsten Forster
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter Ghazal
- Division of Infection and Pathway Medicine, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
180
|
In utero development of memory T cells. Semin Immunopathol 2017; 39:585-592. [DOI: 10.1007/s00281-017-0650-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/07/2017] [Indexed: 12/24/2022]
|
181
|
Sacristán C. Formidable Defenses In Utero. Cell 2017; 170:595-597. [PMID: 28802033 DOI: 10.1016/j.cell.2017.07.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A developing human embryo encounters a multitude of threatening scenarios in the womb. How does the fetus defend itself throughout gestation? A new study by McGovern et al. provides remarkable insight into maternal-fetal immunotolerance.
Collapse
|
182
|
Schliefsteiner C, Peinhaupt M, Kopp S, Lögl J, Lang-Olip I, Hiden U, Heinemann A, Desoye G, Wadsack C. Human Placental Hofbauer Cells Maintain an Anti-inflammatory M2 Phenotype despite the Presence of Gestational Diabetes Mellitus. Front Immunol 2017; 8:888. [PMID: 28824621 PMCID: PMC5534476 DOI: 10.3389/fimmu.2017.00888] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/11/2017] [Indexed: 11/13/2022] Open
Abstract
Background Hofbauer cells (HBCs) are macrophages of the feto-placental unit. Despite the general view that these cells have an anti-inflammatory M2 phenotype, recent studies have claimed that pregnancy pathologies—e.g., gestational diabetes mellitus (GDM)—cause a switch from an M2 to an M1 pro-inflammatory phenotype in HBCs. The pilot-study presented here challenges this claim, showing that HBCs maintain anti-inflammatory properties in spite of the hyperglycemic, low-grade inflammatory environment of GDM. Methods HBCs were isolated from placentae of healthy women (N = 5) and women with GDM (N = 6) diagnosed in the second trimester. FACS was used to measure surface markers associated with either M1 or M2 phenotype on the cells. In addition, placental tissue sections were subjected to immune histochemical imaging to assess the phenotype within the tissue context. Supernatant from control and GDM HBCs was collected at defined time points and used in a multiplex ELISA-on-beads approach to assess secretion of cytokines, chemokines, and growth factors. The effect of HBC cell culture supernatant on placental endothelial activation was investigated. Results FACS and immune staining showed that, indeed, M2 markers, such as CD206 and CD209, are increased in HBCs isolated from GDM placentae. Also, the M1 marker CD86 was increased, but only by trend. Secretion of numerous cytokines, chemokines and growth factors was not changed; pro-inflammatory interleukin (IL)-1β and IL-6 release form GDM HBC was increased but not significant. Exposure to GDM HBC supernatant did not induce cell adhesion molecules (VCAM-1, selectins, vascular endothelial-cadherin) in placental endothelial cells compared to supernatant from control HBCs, an induction of intracellular adhesion molecule 1 was observed however. Conclusion Our study—although performed in a small set of patients—shows that placental macrophages maintain their anti-inflammatory, tissue remodeling M2 phenotype even in pregnancies affected by gestational diabetes. This consistent phenotype might be important for propagation of maternal tolerance toward the fetus and for protection of the fetus from a low-grade inflammatory environment.
Collapse
Affiliation(s)
- Carolin Schliefsteiner
- Perinatal Research Laboratory, Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Miriam Peinhaupt
- Department of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Susanne Kopp
- Perinatal Research Laboratory, Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Jelena Lögl
- Perinatal Research Laboratory, Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria.,Department of Cell Biology, Medical University of Graz, Graz, Austria.,Department of Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Ingrid Lang-Olip
- Department of Cell Biology, Medical University of Graz, Graz, Austria.,Department of Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Ursula Hiden
- Perinatal Research Laboratory, Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Department of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Gernot Desoye
- Perinatal Research Laboratory, Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Christian Wadsack
- Perinatal Research Laboratory, Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| |
Collapse
|
183
|
Fetal dendritic cells give mum a break. Immunol Cell Biol 2017; 95:575-576. [PMID: 28741621 DOI: 10.1038/icb.2017.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
184
|
Early life immunology: Fetal DCs - born to be mild. Nat Rev Immunol 2017; 17:465. [PMID: 28690316 DOI: 10.1038/nri.2017.79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
185
|
VanHook AM. Papers of note in
Nature
546
(7660). Sci Signal 2017. [DOI: 10.1126/scisignal.aao2378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
This week’s articles highlight a mechanism by which melanoma cells prepare the metastatic niche, intestinal defense against rotavirus, and immune tolerance in the human fetus.
Collapse
|