Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Nature ; 607(7919): 540-547, 2022 07.
Article in English | MEDLINE | ID: mdl-35794482

ABSTRACT

Gonadal development is a complex process that involves sex determination followed by divergent maturation into either testes or ovaries1. Historically, limited tissue accessibility, a lack of reliable in vitro models and critical differences between humans and mice have hampered our knowledge of human gonadogenesis, despite its importance in gonadal conditions and infertility. Here, we generated a comprehensive map of first- and second-trimester human gonads using a combination of single-cell and spatial transcriptomics, chromatin accessibility assays and fluorescent microscopy. We extracted human-specific regulatory programmes that control the development of germline and somatic cell lineages by profiling equivalent developmental stages in mice. In both species, we define the somatic cell states present at the time of sex specification, including the bipotent early supporting population that, in males, upregulates the testis-determining factor SRY and sPAX8s, a gonadal lineage located at the gonadal-mesonephric interface. In females, we resolve the cellular and molecular events that give rise to the first and second waves of granulosa cells that compartmentalize the developing ovary to modulate germ cell differentiation. In males, we identify human SIGLEC15+ and TREM2+ fetal testicular macrophages, which signal to somatic cells outside and inside the developing testis cords, respectively. This study provides a comprehensive spatiotemporal map of human and mouse gonadal differentiation, which can guide in vitro gonadogenesis.


Subject(s)
Cell Lineage , Germ Cells , Ovary , Sex Differentiation , Single-Cell Analysis , Testis , Animals , Chromatin/genetics , Chromatin/metabolism , Female , Germ Cells/cytology , Germ Cells/metabolism , Granulosa Cells/cytology , Granulosa Cells/metabolism , Humans , Immunoglobulins , Macrophages/metabolism , Male , Membrane Glycoproteins , Membrane Proteins , Mice , Microscopy, Fluorescence , Ovary/cytology , Ovary/embryology , PAX8 Transcription Factor , Pregnancy , Pregnancy Trimester, First , Pregnancy Trimester, Second , Receptors, Immunologic , Sex Differentiation/genetics , Testis/cytology , Testis/embryology , Transcriptome
2.
Nature ; 597(7875): 250-255, 2021 09.
Article in English | MEDLINE | ID: mdl-34497389

ABSTRACT

The cellular landscape of the human intestinal tract is dynamic throughout life, developing in utero and changing in response to functional requirements and environmental exposures. Here, to comprehensively map cell lineages, we use single-cell RNA sequencing and antigen receptor analysis of almost half a million cells from up to 5 anatomical regions in the developing and up to 11 distinct anatomical regions in the healthy paediatric and adult human gut. This reveals the existence of transcriptionally distinct BEST4 epithelial cells throughout the human intestinal tract. Furthermore, we implicate IgG sensing as a function of intestinal tuft cells. We describe neural cell populations in the developing enteric nervous system, and predict cell-type-specific expression of genes associated with Hirschsprung's disease. Finally, using a systems approach, we identify key cell players that drive the formation of secondary lymphoid tissue in early human development. We show that these programs are adopted in inflammatory bowel disease to recruit and retain immune cells at the site of inflammation. This catalogue of intestinal cells will provide new insights into cellular programs in development, homeostasis and disease.


Subject(s)
Aging , Enteric Nervous System/cytology , Fetus/cytology , Health , Intestines/cytology , Intestines/growth & development , Lymph Nodes/cytology , Lymph Nodes/growth & development , Adult , Animals , Child , Crohn Disease/pathology , Datasets as Topic , Enteric Nervous System/anatomy & histology , Enteric Nervous System/embryology , Enteric Nervous System/growth & development , Epithelial Cells/cytology , Female , Fetus/anatomy & histology , Fetus/embryology , Humans , Intestines/embryology , Intestines/innervation , Lymph Nodes/embryology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Organogenesis , Receptors, IgG/metabolism , Signal Transduction , Spatio-Temporal Analysis , Time Factors
3.
Nature ; 598(7880): 327-331, 2021 10.
Article in English | MEDLINE | ID: mdl-34588693

ABSTRACT

Haematopoiesis in the bone marrow (BM) maintains blood and immune cell production throughout postnatal life. Haematopoiesis first emerges in human BM at 11-12 weeks after conception1,2, yet almost nothing is known about how fetal BM (FBM) evolves to meet the highly specialized needs of the fetus and newborn. Here we detail the development of FBM, including stroma, using multi-omic assessment of mRNA and multiplexed protein epitope expression. We find that the full blood and immune cell repertoire is established in FBM in a short time window of 6-7 weeks early in the second trimester. FBM promotes rapid and extensive diversification of myeloid cells, with granulocytes, eosinophils and dendritic cell subsets emerging for the first time. The substantial expansion of B lymphocytes in FBM contrasts with fetal liver at the same gestational age. Haematopoietic progenitors from fetal liver, FBM and cord blood exhibit transcriptional and functional differences that contribute to tissue-specific identity and cellular diversification. Endothelial cell types form distinct vascular structures that we show are regionally compartmentalized within FBM. Finally, we reveal selective disruption of B lymphocyte, erythroid and myeloid development owing to a cell-intrinsic differentiation bias as well as extrinsic regulation through an altered microenvironment in Down syndrome (trisomy 21).


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow , Down Syndrome/blood , Down Syndrome/immunology , Fetus/cytology , Hematopoiesis , Immune System/cytology , B-Lymphocytes/cytology , Dendritic Cells/cytology , Down Syndrome/metabolism , Down Syndrome/pathology , Endothelial Cells/pathology , Eosinophils/cytology , Erythroid Cells/cytology , Granulocytes/cytology , Humans , Immunity , Myeloid Cells/cytology , Stromal Cells/cytology
4.
Nature ; 574(7778): 365-371, 2019 10.
Article in English | MEDLINE | ID: mdl-31597962

ABSTRACT

Definitive haematopoiesis in the fetal liver supports self-renewal and differentiation of haematopoietic stem cells and multipotent progenitors (HSC/MPPs) but remains poorly defined in humans. Here, using single-cell transcriptome profiling of approximately 140,000 liver and 74,000 skin, kidney and yolk sac cells, we identify the repertoire of human blood and immune cells during development. We infer differentiation trajectories from HSC/MPPs and evaluate the influence of the tissue microenvironment on blood and immune cell development. We reveal physiological erythropoiesis in fetal skin and the presence of mast cells, natural killer and innate lymphoid cell precursors in the yolk sac. We demonstrate a shift in the haemopoietic composition of fetal liver during gestation away from being predominantly erythroid, accompanied by a parallel change in differentiation potential of HSC/MPPs, which we functionally validate. Our integrated map of fetal liver haematopoiesis provides a blueprint for the study of paediatric blood and immune disorders, and a reference for harnessing the therapeutic potential of HSC/MPPs.


Subject(s)
Fetus/cytology , Hematopoiesis , Liver/cytology , Liver/embryology , Blood Cells/cytology , Cellular Microenvironment , Female , Fetus/metabolism , Flow Cytometry , Gene Expression Profiling , Humans , Liver/metabolism , Lymphoid Tissue/cytology , Single-Cell Analysis , Stem Cells/metabolism
5.
Nature ; 563(7731): 347-353, 2018 11.
Article in English | MEDLINE | ID: mdl-30429548

ABSTRACT

During early human pregnancy the uterine mucosa transforms into the decidua, into which the fetal placenta implants and where placental trophoblast cells intermingle and communicate with maternal cells. Trophoblast-decidual interactions underlie common diseases of pregnancy, including pre-eclampsia and stillbirth. Here we profile the transcriptomes of about 70,000 single cells from first-trimester placentas with matched maternal blood and decidual cells. The cellular composition of human decidua reveals subsets of perivascular and stromal cells that are located in distinct decidual layers. There are three major subsets of decidual natural killer cells that have distinctive immunomodulatory and chemokine profiles. We develop a repository of ligand-receptor complexes and a statistical tool to predict the cell-type specificity of cell-cell communication via these molecular interactions. Our data identify many regulatory interactions that prevent harmful innate or adaptive immune responses in this environment. Our single-cell atlas of the maternal-fetal interface reveals the cellular organization of the decidua and placenta, and the interactions that are critical for placentation and reproductive success.


Subject(s)
Cell Communication , Fetus/cytology , Histocompatibility, Maternal-Fetal/immunology , Placenta/cytology , Placenta/metabolism , Pregnancy/immunology , Single-Cell Analysis , Cell Communication/immunology , Cell Differentiation/genetics , Decidua/cytology , Decidua/immunology , Decidua/metabolism , Female , Fetus/immunology , Fetus/metabolism , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Ligands , Placenta/immunology , RNA, Small Cytoplasmic/genetics , Sequence Analysis, RNA , Stromal Cells/cytology , Stromal Cells/metabolism , Transcriptome , Trophoblasts/cytology , Trophoblasts/immunology , Trophoblasts/metabolism
6.
J Virol ; 91(20)2017 10 15.
Article in English | MEDLINE | ID: mdl-28768867

ABSTRACT

Viruses manipulate the complex interferon and interferon-stimulated gene (ISG) system in different ways. We have previously shown that HIV inhibits type I and III interferons in its key target cells but directly stimulates a subset of >20 ISGs in macrophages and dendritic cells, many of which are antiviral. Here, we examine the mechanism of induction of ISGs and show this occurs in two phases. The first phase was transient (0 to 24 h postinfection [hpi]), induced mainly by extracellular vesicles and one of its component proteins, HSP90α, contained within the HIV inoculum. The second, dominant, and persistent phase (>48 hpi) was induced via newly transcribed HIV RNA and sensed via RIGI, as shown by the reduction in ISG expression after the knockdown of the RIGI adaptor, MAVS, by small interfering RNA (siRNA) and the inhibition of both the initiation and elongation of HIV transcription by short hairpin RNA (shRNA) transcriptional silencing. We further define the induction pathway, showing sequential HIV RNA stimulation via Tat, RIGI, MAVS, IRF1, and IRF7, also identified by siRNA knockdown. IRF1 also plays a key role in the first phase. We also show that the ISGs IFIT1 to -3 inhibit HIV production, measured as extracellular infectious virus. All induced antiviral ISGs probably lead to restriction of HIV replication in macrophages, contributing to a persistent, noncytopathic infection, while the inhibition of interferon facilitates spread to adjacent cells. Both may influence the size of macrophage HIV reservoirs in vivo Elucidating the mechanisms of ISG induction may help in devising immunotherapeutic strategies to limit the size of these reservoirs.IMPORTANCE HIV, like other viruses, manipulates the antiviral interferon and interferon-stimulated gene (ISG) system to facilitate its initial infection and establishment of viral reservoirs. HIV specifically inhibits all type I and III interferons in its target cells, including macrophages, dendritic cells, and T cells. It also induces a subset of over 20 ISGs of differing compositions in each cell target. This occurs in two temporal phases in macrophages. Extracellular vesicles contained within the inoculum induce the first, transient phase of ISGs. Newly transcribed HIV RNA induce the second, dominant ISG phase, and here, the full induction pathway is defined. Therefore, HIV nucleic acids, which are potent inducers of interferon and ISGs, are initially concealed, and antiviral ISGs are not fully induced until replication is well established. These antiviral ISGs may contribute to persistent infection in macrophages and to the establishment of viral reservoirs in vivo.


Subject(s)
Gene Expression Regulation , HIV-1/physiology , Interferons/metabolism , Macrophages/virology , RNA, Viral/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Dendritic Cells/virology , HSP90 Heat-Shock Proteins/metabolism , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , RNA, Small Interfering , RNA-Binding Proteins , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Signal Transduction
7.
Rev Med Virol ; 27(2)2017 03.
Article in English | MEDLINE | ID: mdl-28044388

ABSTRACT

Langerhans cells (LCs) situated in stratified squamous epithelium of the skin and mucosal tissue are amongst the first cells that sexually transmitted pathogens encounter during transmission. They are potent antigen presenting cells and play a key role in the host mounting an appropriate immune response. As such, viruses have evolved complex strategies to manipulate these cells to facilitate successful transmission. One of best studied examples is HIV, which manipulates the natural function of these cells to interact with CD4 T cells, which are the main target cell for HIV in which rapid replication occurs. However, there is controversy in the literature as to the role that LCs play in this process. Langerhans cells also play a key role in the way the body mounts an immune response to HSV, and there is also a complex interplay between the transmission of HSV and HIV that involves LCs. In this article, we review both past and present literatures with a particular focus on a few very recent studies that shed new light on the role that LCs play in the transmission and immune response to these 2 pathogens.


Subject(s)
HIV Infections/transmission , Herpes Genitalis/transmission , Host-Pathogen Interactions , Langerhans Cells/immunology , Langerhans Cells/virology , Humans
8.
J Virol ; 89(13): 6575-84, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25855743

ABSTRACT

UNLABELLED: Dendritic cells (DCs) and macrophages are present in the tissues of the anogenital tract, where HIV-1 transmission occurs in almost all cases. These cells are both target cells for HIV-1 and represent the first opportunity for the virus to interfere with innate recognition. Previously we have shown that both cell types fail to produce type I interferons (IFNs) in response to HIV-1 but that, unlike T cells, the virus does not block IFN induction by targeting IFN regulatory factor 3 (IRF3) for cellular degradation. Thus, either HIV-1 inhibits IFN induction by an alternate mechanism or, less likely, these cells fail to sense HIV-1. Here we show that HIV-1 (but not herpes simplex virus 2 [HSV-2] or Sendai virus)-exposed DCs and macrophages fail to induce the expression of all known type I and III IFN genes. These cells do sense the virus, and pattern recognition receptor (PRR)-induced signaling pathways are triggered. The precise stage in the IFN-inducing signaling pathway that HIV-1 targets to block IFN induction was identified; phosphorylation but not K63 polyubiquitination of TANK-binding kinase 1 (TBK1) was completely inhibited. Two HIV-1 accessory proteins, Vpr and Vif, were shown to bind to TBK1, and their individual deletion partly restored IFN-ß expression. Thus, the inhibition of TBK1 autophosphorylation by binding of these proteins appears to be the principal mechanism by which HIV-1 blocks type I and III IFN induction in myeloid cells. IMPORTANCE: Dendritic cells (DCs) and macrophages are key HIV target cells. Therefore, definition of how HIV impairs innate immune responses to initially establish infection is essential to design preventative interventions, especially by restoring initial interferon production. Here we demonstrate how HIV-1 blocks interferon induction by inhibiting the function of a key kinase in the interferon signaling pathway, TBK1, via two different viral accessory proteins. Other viral proteins have been shown to target the general effects of TBK1, but this precise targeting between ubiquitination and phosphorylation of TBK1 is novel.


Subject(s)
Dendritic Cells/immunology , HIV-1/immunology , Host-Pathogen Interactions , Macrophages/immunology , Protein Serine-Threonine Kinases/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Cells, Cultured , Dendritic Cells/virology , Humans , Immune Evasion , Interferons/antagonists & inhibitors , Macrophages/virology , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction , Ubiquitination
9.
J Immunol ; 193(5): 2554-64, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25070850

ABSTRACT

Epidermal Langerhans cells (eLCs) uniquely express the C-type lectin receptor langerin in addition to the HIV entry receptors CD4 and CCR5. They are among the first target cells to encounter HIV in the anogenital stratified squamous mucosa during sexual transmission. Previous reports on the mechanism of HIV transfer to T cells and the role of langerin have been contradictory. In this study, we examined HIV replication and langerin-mediated viral transfer by authentic immature eLCs and model Mutz-3 LCs. eLCs were productively infected with HIV, whereas Mutz-3 LCs were not susceptible because of a lack of CCR5 expression. Two successive phases of HIV viral transfer to T cells via cave/vesicular trafficking and de novo replication were observed with eLCs as previously described in monocyte-derived or blood dendritic cells, but only first phase transfer was observed with Mutz-3 LCs. Langerin was expressed as trimers after cross-linking on the cell surface of Mutz-3 LCs and in this form preferentially bound HIV envelope protein gp140 and whole HIV particles via the carbohydrate recognition domain (CRD). Both phases of HIV transfer from eLCs to T cells were inhibited when eLCs were pretreated with a mAb to langerin CRD or when HIV was pretreated with a soluble langerin trimeric extracellular domain or by a CRD homolog. However, the langerin homolog did not inhibit direct HIV infection of T cells. These two novel soluble langerin inhibitors could be developed to prevent HIV uptake, infection, and subsequent transfer to T cells during early stages of infection.


Subject(s)
Antigens, CD/immunology , HIV Infections/immunology , HIV-1/physiology , Langerhans Cells/immunology , Lectins, C-Type/immunology , Mannose-Binding Lectins/immunology , T-Lymphocytes/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Biological Transport/immunology , HIV Infections/pathology , Humans , Langerhans Cells/pathology , Langerhans Cells/virology , Lectins, C-Type/antagonists & inhibitors , Mannose-Binding Lectins/antagonists & inhibitors , T-Lymphocytes/pathology , T-Lymphocytes/virology , Virus Replication
10.
PLoS Pathog ; 9(10): e1003700, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204260

ABSTRACT

HIV-1 is taken up by immature monocyte derived dendritic cells (iMDDCs) into tetraspanin rich caves from which the virus can either be transferred to T lymphocytes or enter into endosomes resulting in degradation. HIV-1 binding and fusion with the DC membrane results in low level de novo infection that can also be transferred to T lymphocytes at a later stage. We have previously reported that HIV-1 can induce partial maturation of iMDDCs at both stages of trafficking. Here we show that CD45⁺ microvesicles (MV) which contaminate purified HIV-1 inocula due to similar size and density, affect DC maturation, de novo HIV-1 infection and transfer to T lymphocytes. Comparing iMDDCs infected with CD45-depleted HIV-1BaL or matched non-depleted preparations, the presence of CD45⁺ MVs was shown to enhance DC maturation and ICAM-1 (CD54) expression, which is involved in DC∶T lymphocyte interactions, while restricting HIV-1 infection of MDDCs. Furthermore, in the DC culture HIV-1 infected (p24⁺) MDDCs were more mature than bystander cells. Depletion of MVs from the HIV-1 inoculum markedly inhibited DC∶T lymphocyte clustering and the induction of alloproliferation as well as limiting HIV-1 transfer from DCs to T lymphocytes. The effects of MV depletion on these functions were reversed by the re-addition of purified MVs from activated but not non-activated SUPT1.CCR5-CL.30 or primary T cells. Analysis of the protein complement of these MVs and of these HIV-1 inocula before and after MV depletion showed that Heat Shock Proteins (HSPs) and nef were the likely DC maturation candidates. Recombinant HSP90α and ß and nef all induced DC maturation and ICAM-1 expression, greater when combined. These results suggest that MVs contaminating HIV-1 released from infected T lymphocytes may be biologically important, especially in enhancing T cell activation, during uptake by DCs in vitro and in vivo, particularly as MVs have been detected in the circulation of HIV-1 infected subjects.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , HIV Infections/immunology , HIV-1/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , Cell Adhesion/immunology , Cells, Cultured , Dendritic Cells/pathology , HIV Infections/pathology , Humans , Monocytes/immunology , Monocytes/pathology , T-Lymphocytes/pathology
11.
J Immunol ; 190(1): 66-79, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23183897

ABSTRACT

The lineage relationships and fate of human dendritic cells (DCs) have significance for a number of diseases including HIV where both blood and tissue DCs may be infected. We used gene expression profiling of human monocyte and DC subpopulations sorted directly from blood and skin to define the lineage relationships. We also compared these with monocyte-derived DCs (MDDCs) and MUTZ3 Langerhans cells (LCs) to investigate their relevance as model skin DCs. Hierarchical clustering analysis showed that myeloid DCs clustered according to anatomical origin rather than putative lineage. Plasmacytoid DCs formed the most discrete cluster, but ex vivo myeloid cells formed separate clusters of cells both in blood and in skin. Separate and specific DC populations could be determined within skin, and the proportion of CD14(+) dermal DCs (DDCs) was reduced and CD1a(+) DDCs increased during culture, suggesting conversion to CD1a(+)-expressing cells in situ. This is consistent with origin of the CD1a(+) DDCs from a local precursor rather than directly from circulating blood DCs or monocyte precursors. Consistent with their use as model skin DCs, the in vitro-derived MDDC and MUTZ3 LC populations grouped within the skin DC cluster. MDDCs clustered most closely to CD14(+) DDCs; furthermore, common unique patterns of C-type lectin receptor expression were identified between these two cell types. MUTZ3 LCs, however, did not cluster closely with ex vivo-derived LCs. We identified differential expression of novel genes in monocyte and DC subsets including genes related to DC surface receptors (including C-type lectin receptors, TLRs, and galectins).


Subject(s)
Cell Lineage/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dermis/cytology , Dermis/immunology , Epidermal Cells , Epidermis/immunology , Biomarkers/blood , Biomarkers/metabolism , Cell Line, Tumor , Cells, Cultured , Dendritic Cells/pathology , Dermis/pathology , Epidermis/pathology , Galectins/biosynthesis , Galectins/blood , Galectins/physiology , Humans , Langerhans Cells/immunology , Langerhans Cells/metabolism , Langerhans Cells/pathology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Monocytes/cytology , Monocytes/immunology , Monocytes/pathology
12.
Science ; 381(6659): eadd7564, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37590359

ABSTRACT

The extraembryonic yolk sac (YS) ensures delivery of nutritional support and oxygen to the developing embryo but remains ill-defined in humans. We therefore assembled a comprehensive multiomic reference of the human YS from 3 to 8 postconception weeks by integrating single-cell protein and gene expression data. Beyond its recognized role as a site of hematopoiesis, we highlight roles in metabolism, coagulation, vascular development, and hematopoietic regulation. We reconstructed the emergence and decline of YS hematopoietic stem and progenitor cells from hemogenic endothelium and revealed a YS-specific accelerated route to macrophage production that seeds developing organs. The multiorgan functions of the YS are superseded as intraembryonic organs develop, effecting a multifaceted relay of vital functions as pregnancy proceeds.


Subject(s)
Embryonic Development , Yolk Sac , Female , Humans , Pregnancy , Blood Coagulation/genetics , Macrophages , Yolk Sac/cytology , Yolk Sac/metabolism , Embryonic Development/genetics , Atlases as Topic , Gene Expression , Gene Expression Profiling , Hematopoiesis/genetics , Liver/embryology
13.
Science ; 376(6597): eabo0510, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35549310

ABSTRACT

Single-cell genomics studies have decoded the immune cell composition of several human prenatal organs but were limited in describing the developing immune system as a distributed network across tissues. We profiled nine prenatal tissues combining single-cell RNA sequencing, antigen-receptor sequencing, and spatial transcriptomics to reconstruct the developing human immune system. This revealed the late acquisition of immune-effector functions by myeloid and lymphoid cell subsets and the maturation of monocytes and T cells before peripheral tissue seeding. Moreover, we uncovered system-wide blood and immune cell development beyond primary hematopoietic organs, characterized human prenatal B1 cells, and shed light on the origin of unconventional T cells. Our atlas provides both valuable data resources and biological insights that will facilitate cell engineering, regenerative medicine, and disease understanding.


Subject(s)
Immune System , Lymphocytes , Monocytes , Genomics , Humans , Immune System/embryology , Lymphocytes/metabolism , Monocytes/metabolism , Organ Specificity , RNA-Seq , Single-Cell Analysis
14.
Nat Cell Biol ; 24(10): 1487-1498, 2022 10.
Article in English | MEDLINE | ID: mdl-36109670

ABSTRACT

The liver has been studied extensively due to the broad number of diseases affecting its vital functions. However, therapeutic advances have been hampered by the lack of knowledge concerning human hepatic development. Here, we addressed this limitation by describing the developmental trajectories of different cell types that make up the human liver at single-cell resolution. These transcriptomic analyses revealed that sequential cell-to-cell interactions direct functional maturation of hepatocytes, with non-parenchymal cells playing essential roles during organogenesis. We utilized this information to derive bipotential hepatoblast organoids and then exploited this model system to validate the importance of signalling pathways in hepatocyte and cholangiocyte specification. Further insights into hepatic maturation also enabled the identification of stage-specific transcription factors to improve the functionality of hepatocyte-like cells generated from human pluripotent stem cells. Thus, our study establishes a platform to investigate the basic mechanisms directing human liver development and to produce cell types for clinical applications.


Subject(s)
Hepatocytes , Liver , Humans , Liver/metabolism , Hepatocytes/metabolism , Cell Differentiation , Organoids , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Genome Biol ; 22(1): 346, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34930412

ABSTRACT

Multimodal data is rapidly growing in many fields of science and engineering, including single-cell biology. We introduce MultiMAP, a novel algorithm for dimensionality reduction and integration. MultiMAP can integrate any number of datasets, leverages features not present in all datasets, is not restricted to a linear mapping, allows the user to specify the influence of each dataset, and is extremely scalable to large datasets. We apply MultiMAP to single-cell transcriptomics, chromatin accessibility, methylation, and spatial data and show that it outperforms current approaches. On a new thymus dataset, we use MultiMAP to integrate cells along a temporal trajectory. This enables quantitative comparison of transcription factor expression and binding site accessibility over the course of T cell differentiation, revealing patterns of expression versus binding site opening kinetics.


Subject(s)
Chromosome Mapping/methods , Single-Cell Analysis/methods , Transcriptome , Algorithms , Chromatin , Chromosomes, Human , Gene Expression Regulation , Genetic Markers , Genomics , Humans , Software , Transcription Factors
16.
Nat Commun ; 12(1): 2147, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846309

ABSTRACT

Tissue mononuclear phagocytes (MNP) are specialised in pathogen detection and antigen presentation. As such they deliver HIV to its primary target cells; CD4 T cells. Most MNP HIV transmission studies have focused on epithelial MNPs. However, as mucosal trauma and inflammation are now known to be strongly associated with HIV transmission, here we examine the role of sub-epithelial MNPs which are present in a diverse array of subsets. We show that HIV can penetrate the epithelial surface to interact with sub-epithelial resident MNPs in anogenital explants and define the full array of subsets that are present in the human anogenital and colorectal tissues that HIV may encounter during sexual transmission. In doing so we identify two subsets that preferentially take up HIV, become infected and transmit the virus to CD4 T cells; CD14+CD1c+ monocyte-derived dendritic cells and langerin-expressing conventional dendritic cells 2 (cDC2).


Subject(s)
Anal Canal/cytology , Antigens, CD/metabolism , Dendritic Cells/metabolism , Genitalia/cytology , HIV-1/physiology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Monocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Shape , Collagenases/metabolism , Dermis/metabolism , HIV Infections/immunology , HIV Infections/virology , Humans , Lipopolysaccharide Receptors/metabolism , Mucous Membrane/metabolism , Phagocytes/metabolism , Phenotype , Receptors, CCR5/metabolism , Sialic Acid Binding Ig-like Lectin 1/metabolism , Transcription, Genetic
17.
Nat Commun ; 12(1): 7092, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34876592

ABSTRACT

The nasal epithelium is a plausible entry point for SARS-CoV-2, a site of pathogenesis and transmission, and may initiate the host response to SARS-CoV-2. Antiviral interferon (IFN) responses are critical to outcome of SARS-CoV-2. Yet little is known about the interaction between SARS-CoV-2 and innate immunity in this tissue. Here we apply single-cell RNA sequencing and proteomics to a primary cell model of human nasal epithelium differentiated at air-liquid interface. SARS-CoV-2 demonstrates widespread tropism for nasal epithelial cell types. The host response is dominated by type I and III IFNs and interferon-stimulated gene products. This response is notably delayed in onset relative to viral gene expression and compared to other respiratory viruses. Nevertheless, once established, the paracrine IFN response begins to impact on SARS-CoV-2 replication. When provided prior to infection, recombinant IFNß or IFNλ1 induces an efficient antiviral state that potently restricts SARS-CoV-2 viral replication, preserving epithelial barrier integrity. These data imply that the IFN-I/III response to SARS-CoV-2 initiates in the nasal airway and suggest nasal delivery of recombinant IFNs to be a potential chemoprophylactic strategy.


Subject(s)
Epithelial Cells/virology , Interferon Type I/immunology , Interferons/immunology , Nasal Mucosa/virology , SARS-CoV-2/physiology , Antiviral Agents/immunology , Antiviral Agents/pharmacology , COVID-19/immunology , COVID-19/virology , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/immunology , Humans , Immunity, Innate , Kinetics , Nasal Mucosa/cytology , Nasal Mucosa/immunology , SARS-CoV-2/drug effects , Signal Transduction/drug effects , Viral Tropism , Virus Replication/drug effects , Interferon Lambda
18.
Science ; 371(6527)2021 01 22.
Article in English | MEDLINE | ID: mdl-33479125

ABSTRACT

The skin confers biophysical and immunological protection through a complex cellular network established early in embryonic development. We profiled the transcriptomes of more than 500,000 single cells from developing human fetal skin, healthy adult skin, and adult skin with atopic dermatitis and psoriasis. We leveraged these datasets to compare cell states across development, homeostasis, and disease. Our analysis revealed an enrichment of innate immune cells in skin during the first trimester and clonal expansion of disease-associated lymphocytes in atopic dermatitis and psoriasis. We uncovered and validated in situ a reemergence of prenatal vascular endothelial cell and macrophage cellular programs in atopic dermatitis and psoriasis lesional skin. These data illustrate the dynamism of cutaneous immunity and provide opportunities for targeting pathological developmental programs in inflammatory skin diseases.


Subject(s)
Dermatitis, Atopic/embryology , Dermatitis, Atopic/pathology , Psoriasis/embryology , Psoriasis/pathology , Skin/embryology , Animals , Atlases as Topic , Cell Movement , Datasets as Topic , Dendritic Cells/immunology , Dermatitis, Atopic/immunology , Dermatologic Agents/pharmacology , Humans , Immunity, Innate/genetics , Methotrexate/pharmacology , Mice , Phagocytes/immunology , Psoriasis/immunology , Single-Cell Analysis , Skin/cytology , Skin/immunology , T-Lymphocytes/immunology , Transcriptome
19.
Nat Med ; 27(5): 904-916, 2021 05.
Article in English | MEDLINE | ID: mdl-33879890

ABSTRACT

Analysis of human blood immune cells provides insights into the coordinated response to viral infections such as severe acute respiratory syndrome coronavirus 2, which causes coronavirus disease 2019 (COVID-19). We performed single-cell transcriptome, surface proteome and T and B lymphocyte antigen receptor analyses of over 780,000 peripheral blood mononuclear cells from a cross-sectional cohort of 130 patients with varying severities of COVID-19. We identified expansion of nonclassical monocytes expressing complement transcripts (CD16+C1QA/B/C+) that sequester platelets and were predicted to replenish the alveolar macrophage pool in COVID-19. Early, uncommitted CD34+ hematopoietic stem/progenitor cells were primed toward megakaryopoiesis, accompanied by expanded megakaryocyte-committed progenitors and increased platelet activation. Clonally expanded CD8+ T cells and an increased ratio of CD8+ effector T cells to effector memory T cells characterized severe disease, while circulating follicular helper T cells accompanied mild disease. We observed a relative loss of IgA2 in symptomatic disease despite an overall expansion of plasmablasts and plasma cells. Our study highlights the coordinated immune response that contributes to COVID-19 pathogenesis and reveals discrete cellular components that can be targeted for therapy.


Subject(s)
COVID-19/immunology , Proteome , SARS-CoV-2/immunology , Single-Cell Analysis/methods , Transcriptome , Cross-Sectional Studies , Humans , Monocytes/immunology , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology
20.
Science ; 367(6480)2020 02 21.
Article in English | MEDLINE | ID: mdl-32079746

ABSTRACT

The thymus provides a nurturing environment for the differentiation and selection of T cells, a process orchestrated by their interaction with multiple thymic cell types. We used single-cell RNA sequencing to create a cell census of the human thymus across the life span and to reconstruct T cell differentiation trajectories and T cell receptor (TCR) recombination kinetics. Using this approach, we identified and located in situ CD8αα+ T cell populations, thymic fibroblast subtypes, and activated dendritic cell states. In addition, we reveal a bias in TCR recombination and selection, which is attributed to genomic position and the kinetics of lineage commitment. Taken together, our data provide a comprehensive atlas of the human thymus across the life span with new insights into human T cell development.


Subject(s)
Atlases as Topic , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Thymus Gland/growth & development , Thymus Gland/immunology , CD8-Positive T-Lymphocytes/cytology , Dendritic Cells/cytology , Dendritic Cells/immunology , Fibroblasts/cytology , Fibroblasts/immunology , Humans , RNA-Seq/methods , Receptors, Antigen, T-Cell/metabolism , Single-Cell Analysis/methods , Thymus Gland/cytology
SELECTION OF CITATIONS
SEARCH DETAIL